EP3997103A1 - Compounds comprising a fibroblast activation protein ligand and use thereof - Google Patents

Compounds comprising a fibroblast activation protein ligand and use thereof

Info

Publication number
EP3997103A1
EP3997103A1 EP20735646.0A EP20735646A EP3997103A1 EP 3997103 A1 EP3997103 A1 EP 3997103A1 EP 20735646 A EP20735646 A EP 20735646A EP 3997103 A1 EP3997103 A1 EP 3997103A1
Authority
EP
European Patent Office
Prior art keywords
compound
disease
nos
cys
pro
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP20735646.0A
Other languages
German (de)
English (en)
French (fr)
Inventor
Frank Osterkamp
Dirk Zboralski
Eberhard Schneider
Christian Haase
Matthias Paschke
Aileen Höhne
Jan Ungewiß
Christiane Smerling
Ulrich Reineke
Anne BREDENBECK
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
3B Pharmaceuticals GmbH
Original Assignee
3B Pharmaceuticals GmbH
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from EP19000325.1A external-priority patent/EP3763726A1/en
Application filed by 3B Pharmaceuticals GmbH filed Critical 3B Pharmaceuticals GmbH
Publication of EP3997103A1 publication Critical patent/EP3997103A1/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/088Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins conjugates with carriers being peptides, polyamino acids or proteins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention is related to a chemical compound; an inhibitor of fibroblast activation protein (FAP); a composition comprising the compound and inhibitor, respectively; the compound, the inhibitor and the composition, respectively, for use in a method for the diagnosis of a disease; the compound, the inhibitor and the composition, respectively, for use in a method for the treatment of a disease; the compound, the inhibitor and the composition, respectively, for use in a method of diagnosis and treatment of a disease which is also referred to as “thera(g)nosis” or “thera(g)nostics”; the compound, the inhibitor and the composition, respectively, for use in a method for delivering an effector t ⁇ pressing tissue; a method for the diagnosis of a disease using the compound, the inhibitor composition, respectively; a method for the treatment of a disease using the compound, the inhibitor and the composition, respectively; a method for the diagnosis and treatment of a disease which is also referred to as“thera(g)nosis” or“thera(g)nostics, using the compound, the inhibitor and
  • TME tumor microenvironment
  • CAFs have been described as the source and origin for CAFs, such as e.g. fibroblasts, mesenchymal stem cells, smooth muscle cells, cells of epithelial origin, or endothelial cells (Madar, et al, Trends Mol Med, 2013, 19: 447).
  • CAFs exhibit mesenchymal- like features and often are the dominant cell type within a solid tumor mass.
  • CAFs have attracted increasing attention as a player in tumor progression and homeostasis (Gascard, et al., Genes Dev, 2016, 30: 1002; LeBleu, et al, Dis Model Mech, 2018, 11).
  • CAFs (Shiga, et al., Cancers (Basel), 2015, 7: 2443; Pure, et al., Oncogene, 2018, 37: 4343; Jacob, et al., Curr Mol Med, 2012, 12: 1220). Due to the omnipresence of CAFs and stroma within tumors, FAP was discovered as a suitable marker diagnostics and as a suitable target for radiopharmaceutical therapy (Siveke, JNuclMea A' * 59: , >3 ⁇
  • Fibroblast activation protein a is a type II transmembrane serine protease and a member of the S9 prolyl oligopeptidase family (Park, et al., J Biol Chem, 1999, 274: 36505). The closest family member DPP4 shares 53% homology with FAP. Like othc DPP enzymes (DPP4, DPP7, DPP8, DPP9), FAP has post-proline exopeptidase activity. In addition, FAP possesses endopeptidase activity, similar to prolyl oligopeptidase/endopeptidase (POP/PEEP). The FAP gene is highly conserved across various species. The extracellular domain ofhum shares
  • FAP is a 760 amino acid transmembrane protein composed of a short N-termiiial cytoplasmic tail (6 amino acids), a single transmembrane domain (20 amino acids), and a 734 amino acid extracellular domain (Aertgeerts, et al, J Biol Chem, 2005, 280: 19441).
  • This extracellular domain consists of an eight-bladed b-propeller and an a/b hydrolase domain.
  • the catalytic triad is composed of Ser624, Asp702, and His734 and is located at the interface of the b-propeller and the hydrolase domain.
  • the active site is accessible through a central hole of the b-propeller domain or through a narrow cavity between the b-propeller and the hydrolase dome w nonomers are not active, but form active homodimers as well as heterodimers with DP'' * O nersf et al, Cancer Res, JAiMv 66: 4652).
  • Soluble faomodiiverG 17 AP has also been described (Keane, et al, FEBS Open Bio, 2013, 4: 43; Lee, et aI, BIooi' RiF % 107: 1397). sesses dual enzyme activity (Hamson, et al, Proteomics Clin Appl, 2014, 8: 454).
  • FAP substrates that are cleaved rapidly via its dipeptidyl peptidase activity are neuropeptide Y, Peptide YY, Substance P, and B-type natriuretic peptide.
  • Collagen I and III, FGF21 and oc-aiitiplasiiiin have been shown to be cleaved by the endopeptidase activity of FAP. While FAP is unable to cleave native co pre-digestion by other proteases, such as matrix metalloproteinases, facilitates further c i cleavage by FAP.
  • FAP is transiently expressed during normal development, but only rarely in healthy adult tissues. In transgenic mice, it was demonstrated that FAP is expressed by adipose tissue, skeletal muscle, skin, bone and pancreas (Pure, et ah, Oncogene, 2018, 37: 4343; Roberts, e f 1 Exp Med, 20 A d ⁇ However, a FAP knockout mouse has a healthy phenotype, a, ⁇ ' .”.ting a redundant role under normal conditions (Niedermeyer, et al, Mol Cell Biol, 2000. 20: 1089). At sites of active tissue remodeling, including wound healing, fibrosis, arthritis, atherosclerosis and canc comes highly upregulated in stromal cells (Pure, et al, Oncogene, 2018, 37:
  • Ovary + + FAP positivity increases with tumor stage; negative FAP expression is associated with longer disease-free survival FAP positivity detected in canter ceils in 21% of tumors, stromal positivity in 61%, Another study reported stromal positivity in 92% of cancer tissues with extremely rare FAP expression in malignant ceils; it also reported an association with advanced tumor stage and presence of lymph node metastases, FAP- positive malignant cells are present in malignant pleural and peritoneal effusions; strong _ positivity is associated with worse survival, _
  • Glioma + + FAP expression increased in glioblastoma, highest expression found in the mesenchymal
  • FAP expression in CAFs was shown for almost all carcinomas and sarcomas (Pure, et al, Oncogene , 2018, 37: 4343; Busek, et ol. Front Biosci (Landmark Ed), 21)18, 23: 1933). Furthermore, CAFs are present in hematological malignancies (Raffaghello, et at, Oncotarget, 2015, 6: 2589). Utilization of FAP as a therapeutic target is therefore not limited to certain tumor entities.
  • FAP as well i pressing cells present in the tumor microenvironment significantly influence tumor progression (Hanahan, et al, Cancer 21: 309). Additionally, due to its relatively selective expression in tumors, FAP is regarded as a suitable target for therapeutic and diagnostic agents as described below (Siveke, JNucl Med, 2018, 59: 1412; Christiansen, et al, Neoplasia, 2013, 15: 348; Zf et al, Mol Med Rep, 2015, 11: 3203).
  • FAP was utilized as a therapeutic target in cancer.
  • various strategies have been explored, including e.g. inhibition of FAP enzymatic activity, ablation of FAP-positive cells, or targeted delivery of cytotoxic compounds.
  • WO 2008/116054 disclosed hexapeptide derivatives wherein compounds comprise a C- terminal bis-amino or boronic acid functional group.
  • US 2017/0066800 disclosed pseudopeptide inhibitors, such as M83, effective against These inhibitors were assessed in lung and colon cancer xenografts in immiinodeficient mice. A suppression of tumor growth was observed (Jackson, et al, Neoplasia, 2015, 17: 43). These pseudopeptides inhibit the activity of both prolyl oligopeptidase (POP/PREP) and FAP, thereby excluding their use as specific therapeutic FAP inhibitors.
  • POP/PREP prolyl oligopeptidase
  • FAP both prolyl oligopeptidase
  • US 2008/280856 disclosed a nanomolar boronic acid-based inhibitor.
  • the inhibitor shows a bi specific inhibition of FAP and PREP, thereby excluding their use as specific therapeufi inhibitors.
  • ibitors based on cyclic peptides were disclosed, e.g., in WO 2016/146174 and WO 2006/042282.
  • WO 2006/042282 disclosed polypeptides for treatment of melanoma. In nude mice, inhibition of melanoma growth and melanoma metastasis was shown.
  • WO l ⁇ 68708 disclosed a humanize, ( monoclonal antibody, F19, (Sibrotuzumab). Furthermore, the anti-FAP antibody FI 9 and humanized versions thereof were disclosed in WO 99/57151 and WO 01/68708. Development approaches involved e.g. the generation of high affinity, species cross-reactiw. r 4 s -specific scFvs converted into a bivalent derivative (Brocks, etal, Mol Med, 2001, 7: 461).
  • Sibrotuzumab showed specific tumor enrichment whilst failing to demonstrate measurable therapeutic activity in patients with metastatic colorectal cancer, with only 2 out of 17 patients having stable disease (Hoffaeinz, et al, Onkologie, 2003, 26: 44).
  • This F19 antibody has not been shown to block any cellular or protease functic" 17 AP, which might explain the lack of therapeutic effects 44; Scott, et al, Clin Cancer Res, 2003, 9: 1639).
  • US 2018/022822 disclosed novel molecules specifically binding to human FAP and epitopes thereof, as human-derived antibodies and chimeric antigen receptors (CARs) useful in the treatment of diseases and conditions induced b eatment of mice bearing orthotopic syngeneic MC38 colorectal tumors with an anti-FAP antibody reduced the tumor diameter and number of metastasis.
  • WO 2012/020006 disclosed glycoengineered antibodies that bear modified oligosaccharides in the Fc region. Subsequently, bispecific antibodies ic for FAP and DR5 were developed as subject to WO 2014/161845.
  • WO 2010/036814 disclosed small molecule inhibitors of FAP for use as therapeutic agents through inhibition of FAPs enzyme activity or as radiopharmaceuticals through binding to FAP.
  • WO 2019/083990 disclosed imaging and radiotherapeutic agents based on small molecule FAP- inhibitors described by Jansen et al (Jansen, et al, J Med Chem, 2014, 57: 3053; Jansen, et al, ACS Med Chem Lett, 2013, 4: 491).
  • WO 2011/040972 disclosed high-affinity antibodies recognizing both human and murine FAP antigen as potent radioimmunoconjugates.
  • ESC 11 IgGl induces down modulation and internalization of surface FAP (Fischer, et al, Clin Cancer Res, 2012, 18: 6208).
  • WO 2017/21 1809 disclosed tissue targeting thorium-227 complexes wherein the targeting moiety has specificity for FAP.
  • the long circulation time of antibodies makes them unsuitable for a diagnostic, therapeutic, or theragnostic approach involving radionuclides.
  • FAP has also been described as being involved in other diseases than oncology indications, examples of which are given below.
  • Fibroblast-like synoviocytes in rheumatoid arthritic joints of patients show a significantly increased expression of FAP (Bauer, et al, Arthritis Res Ther, 2006, 8: R171 ; Milner, et al. , Arthritis Res Ther, 2006, 8: R23).
  • stromal cells play an important role in organizing the structure of synovial tissue of joints by producing extracellular matrix components, recruiting infiltrating immune cells and secreting inflammatory mediators.
  • Additioi recognized not only as a marker of activated fibroblasts in the injury response (Tillmanns, et al, Int J Cardiol, 2013, 168: 3926) but also : ⁇ nportant player in the healing process of wounds (Ramirez-Montagut, et al, Oncogene, 2bi ⁇ 4, 23: 5435). Jing et al. demonstrated a time-dependent course of change in FAP expression following bum wounds in rats (Jing, et al, Nan Fang Yi Ke Da Xue Xue Bao, 2013, 33: 615).
  • liver fibrosis correlates with the histological severity of liver disease (Gorrell, etal.,AdvEx fi’- v CHG, 524: 235). Thereto also a promising target in the treatment of liver fibrosis (Lay, et al, Front Biosci (Landmark Ed), 2019, 24: 1).
  • FAP is expressed in arteriosclerotic lesions and upregulated in activated vascular smooth muscle cells (Monslow, et al, Circulation, 2013, 128: A17597). Monslow et al. showed that targeted inhibition of FAP in arteriosclerotic lesions may decrease overall lesion burden, inhibit inflammatory cell homing, and increase lesion stability through its ability to alter lesion architecture by favoring matrix-rich lesions over inflammation. More importantly, most of the arteriosclerotic pathologies share a common pathogenic feature: the rupture of an atherosclerotic plaque inducing arteriosclerotic lesions (Davies, et al, Br Heart J, 1985, 53: 363; Falk, rim J Cardiol, 53: 1 14e).
  • Rupture of the fibrous cap in advanced atherosclerotic plaques is a critical trigger of acute coronary syndromes that may lead to myocardial infarction and sudden cardiac death.
  • One of the key events in promoting plaque instability is the degradation of the fibrous cap, which exposes the underlying thrombogenic plaque core to the bloodstream, thereby causing thrombosis and subsequent vessel occlusion (Farb, et al, Circulation, 1996, 93: 1354; Virmani, et al, J Am Coll Cardiol, 2006, 47: C13).
  • Brokopp et al. showed that mtributes to type I collagen breakdown in fibrous caps (Brokopp, et al , Eur Heart J, 2011, 32: 2713).
  • a radiolabeled tracer was developed and its applicability for atherosclerosis imaging shown (Meletta, et al, Molecules, 2015, 20: 2081).
  • the problem underlying the present invention is the provision of a compound which is suitable as a diagnostic agent and/or a pharmaceutical agent, particularly if conjugated to a diagnostically and/or therapeutically active effector.
  • a further problem underlying the present invention is the provision of a compound which is suitable as a diagnostic agent and/or a pharmaceutical agent, particularly if conjugated to a diagnostically and/or therapeutically active effector, whereby the compound is a potent inhibitc activity; preferably the pIC50 of the compound is equal to or greater than 6.0.
  • a further problem underlying the present invention is the provision of a compound which is suitable as a diagnostic agent and/or a pharmaceutical agent, particularly if conjugated to a diagnostically and/or therapeutically active effector, in the diagnosis and/or therapy of a disease where the diseased cells and/or diseased tissues express FAP.
  • a still further problem underlying the instant invention is the provision of a compound which is suitable for delivering a diagnostically and/or therapeutically effective agent to a diseased cell and/or diseased tissue, respectively, and more particulai expressing diseased cell and/or diseased tissue, preferably the diseased tissue comprises or contains cancer associated fibroblasts.
  • a problem underlying the present invention is the provision of a method for the diagnosis of a disease, of a method for the treatment and/or prevention of a disease, and a method for the combined diagnosis and treatment of a disease; preferably such disease is a disease involving i L’ expressing cells and/or tissues, more particularly a ! to’ expressing diseased cell and/or diseased tissue, preferably the diseased tissue comprises or contains cancer associated fibroblasts.
  • a still further problem underlying the present invention is the provision of a method for the identification of a subject, wherein the subject is likely to respond or likely not to respond to a treatment of a disease, a method for the selection of a subject from a group of subjects, wherein the subject is likely to respond or likely not to respond to a treatment of a disease.
  • a problem underlying the present invention is the provision of a pharmaceutical composition containing a compound having the characteristics as outlined above.
  • a problem underlying the present invention is the provision of a kit which is suitable for use in any of the above methods.
  • a compound which is suitable for delivering a diagnostically and/or therapeutically effective agent to a diseased cell and/or disea sue, respectively, and more particularly a FAP -expressing diseased cell and/or diseased tissue preferably the diseased tissue comprises or contains cancer associated fibroblasts.
  • a method for the diagnosis of a disease, of a method for the treatment and/or prevention of a disease, and a method for the combined diagnosis and treatment of a disease preferably such disease is a disease involving FAP- expressing cells and/or tissues, more particularly a FAP-expressing diseased cell and/or diseased tissue, preferably the diseased tissue comprises or contains cancer associated fibroblasts.
  • a method for the identification of a subject wherein the subject is likely to respond or likely not to respond to a treatment of a disease
  • a method for the selection of a subject from a group of subjects wherein the subject is likely to respond or likely not to respond to a treatment of a disease.
  • a pharmaceutical composition containing a compound having the characteristics as outlined above there is a need for a kit which is suitable for use in any of the above methods. The present invention satisfies these needs.
  • Embodiment 1 A compound selected from the group consisting of compound Bex- h (tMeBn(DOTA-AET))-Pi > ''m » v -Gln-Phe-Cys]-OH (3BP-3554) of the following formula
  • Embodiment 2 The compound of Embodiment 1, wherein the compound is compound
  • Embodiment 3 The compound of Embodiment 1, wherein the compound is compound
  • Embodiment 4 Hex-[Cys(tMeBn(DOTA-PP))-Pro-Pro-Thr-Gln-Phe-Cys]-Asp-NH2 (3BP-3407) of the following formula Embodiment 4.
  • Embodiment 5 The compound of any one of Embodiments 1 to 4, wherein the compound is capable of binding to fibroblast activation protein
  • Embodiment 6 The compound of any one of Embodiments 1 to 5, wherein the compound comprises a diagnostically active nuclide or a therapeutically active nuclide.
  • Embodiment 7 The compound of Embodiment 6, wherein the compound is selected from the group comprising compound Hex-[Cys(tMeBn(InDOTA-PP))-Pro-Pro-Thr-Gln-Phe- Cys]-Asp-NH2 (3 BP-3590) of the following formula compound Hex-[Cys(tMeBn(LuDOTA-PP))-Pro-Pro-Thr-Gln-Phe-Cys]-Asp-NH2 (3BP-3590) of the following formula compound Hex-[Cys(tMeBn(LuDOTA-PP))-Pro-Pro-Thr-Gln-Phe-Cys]-Asp-NH2 (3BP-
  • Embodiment 8 The compound of any one of Embodiments 6 and 7, wherein the diagnostically active nuclide is a diagnostically active radionuclide.
  • Embodiment 9 The compound of Embodiment 8, wherein the diagnostically active radionuclide is selected from the group consisting of 43 Sc, 44 Sc, 51 Mn, 52 MII, M Cu, 67 Ga, 68 Ga, 86 Y, 89 Zr, 94m Tc, 99m Tc, m In, I 52 Tb, 155 Tb, 201 T1, 203 Pb, I8 F, 76 Br, 77 Br, 123 I, I24 I, 125 I, preferably 43 Sc, 44 Sc, 64 Cu, 67 Ga, 68 Ga, 86 Y, 89 Zr, 99,I1 Tc, i In, 152 Tb, 155 Tb, 203 Pb, m F, 76 Br, 7 3 ⁇ 4r, 123 1, 124 1, 125 I and most preferably 64 Cu, 68 Ga, 89 Zr, 99ni Tc, 11 : 18 F, 123 1, and , 24 I.
  • the diagnostically active radionuclide is selected from
  • Embodiment 10 The compound of any one of Embodiments 6 and 7, wherein the therapeutically active nuclide is a therapeutically active radionuclide.
  • Embodiment 1 1. compound of Embodiment 10, wherein the therapeutically active radionuclide is selected from the group consisting of 47 Sc, 67 Cu, 89 Sr, 90 Y, 153 Sm, 149 Tb, 161 Tb, I 77 LU, 186 Re, 188 Re, 2 l2 Pb, 20 BI, 223 Ra, 225 Ac, 226 Th, 227 Th, 131 1, 211 At, preferably 47 Sc, 67 Cu, 90 Y, 177 LU, 188 Re, 212 Pb, 213 Bi, 225 Ac, 227 Th, t31 I, 21 'At and most preferably 90 Y, 177 Lu, 225 Ac, 227 Th, 131 I and 21 'At.
  • the therapeutically active radionuclide is selected from the group consisting of 47 Sc, 67 Cu, 89 Sr, 90 Y, 153 Sm, 149 Tb, 161 Tb, I 77 LU, 186 Re, 188 Re,
  • Embodiment 12 The compound of any one of Embodiments 1 to 11, wherein the compound interacts with a fibroblast activation protein (FAP), preferably with human FAP having an amino acid sequence of SEQ ID NO: 1 or a homolog thereof, wherein the amino acid sequence of the homolog has an identity of at least 85% to the amino acid sequence of SEQ ID NO: 1.
  • FAP fibroblast activation protein
  • Embodiment 13 The compound of Embodiment 12, wherein the compound is an inhibitor of the fibroblast activation protein (FAP).
  • FAP fibroblast activation protein
  • Embodiment 14 The compound of any one of Embodiments 1 to 13, for use in a method for the diagnosis of a disease.
  • Embodiment 15 The compound for use of Embodiment 14, wherein the disease is a disease involving fibroblast activation protein (FAP), preferably upregulated expression of fibroblast activation protein (FAP).
  • FAP fibroblast activation protein
  • FAP upregulated expression of fibroblast activation protein
  • Embodiment 16 The compound for use of any one of Embodiments 14 to 15, wherein the disease involves cells showing upregulated expression of fibroblast activation prof preferably diseased tissue containing cells showing upregulated expression of fibroblast activation protein (FAP), more preferably disease involving tumor associated fibroblasts.
  • FAP fibroblast activation protein
  • Embodiment 17 The compound for use of any one of Embodiments 14 to 16, wherein the disease is a neoplasm, preferably a cancer or tumor.
  • Embodiment 18 The compound for use of Embodiment 17, wherein the neoplasm, cancer, and tumor are each and individually selected from the group comprising a solid tumor, an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangi ocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer, melanoma, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, salivary carcinoma, sarcoma, squamous cell carcinoma, and thyroid cancer.
  • Embodiment 19 Embodiment 19.
  • neoplasm, cancer, and tumor are each and individually selected from the group comprising breast cancer, colorectal cancer, cholangiocarcinoma, head and neck cancer, lung cancer, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, and squamous cell carcinoma.
  • Embodiment 20 The compound for use of any one of Embodiments 14 to 16, wherein the disease is selected from the groups comprising inflammatory disease, cardiovascular disease, autoimmune disease, and fibrotic disease.
  • Embodiment 21 The compound for use of Embodiment 20, wherein the disease is an inflammatory disease.
  • Embodiment 22 The compound for use of Embodiment 21, wherein the disease is atherosclerosis, arthritis, or rheumatoid arthritis.
  • Embodiment 23 The compound for use of Embodiment 20, wherein the disease is a cardiovascular disease.
  • Embodiment 24 The compound for use of Embodiment 23, wherein the disease is a cardiovascular disease involving atherosclerotic plaques.
  • Embodiment 25 The compound for use of Embodiment 24, wherein the disease is an atherosclerotic pathology caused by rapture of plaques, acute coronary syndrome, myocardial infarction, thrombosis, or vessel occlusion.
  • Embodiment 26 The compound for use of Embodiment 20, wherein the disease is a fibrotic disease.
  • Embodiment 27 The compound for use of Embodiment 26, wherein the disease is selected form the group comprising idiopathic pulmonary fibrosis, Crohn’s disease, and liver fibrosis.
  • Embodiment 28 The compound for use of any one of Embodiments 14 to 27, wherein the compound comprises a diagnostically active nuclide, preferably a diagnostically active radionuclide.
  • Embodiment 29 The compound for use of Embodiment 28, wherein the diagnostically active nuclide is selected from the group comprising 43 Sc, 44 Sc, 51 Mn, 52 Mn, 64 Cu, 67 Ga, 68 Ga, 86 Y, 89 Zr, 94m Tc, 99m Tc, 11 'In, 152 Tb, !55 Tb, 201 T1, 203 Pb, 18 F, 76 Br, 77 Br, I23 I, 124 I, 125 I, preferably 43 Sc, 44 Sc, 64 Cu, 67 Ga, 68 Ga, 86 Y, 89 Zr, 99m Tc, 1J 1 ln, ] 52 Tb, 155 Tb, 203 Pb, )8 F, 76 Br, 7 3 ⁇ 4r, 123 I, 124 I, 125 1, and more preferably 64 Cu, 68 Ga, 89 Zr, "“Tc, n i In, , 8 F, 123 1, and 124 I.
  • Embodiment 30 The compound for use of any one of Embodiments 14 to 29, wherein the method for the diagnosis is an imaging method.
  • Embodiment 31 The compound for use of Embodiment 30, wherein the imaging method is selected from the group consisting of scintigraphy, Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET).
  • the imaging method is selected from the group consisting of scintigraphy, Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET).
  • SPECT Single Photon Emission Computed Tomography
  • PET Positron Emission Tomography
  • Embodiment 32 The compound for use of any one of Embodiments 14 to 31, wherein the method comprises the administration of a diagnostically effective amount of the compound to a subject, preferably to a mammal, wherein the mammal is selected from the group comprising man, companion animals, pets, and livestock, more preferably the subject is selected from the group comprising man, dog, cat, horse, and cow, and most preferably the subject is a human being.
  • Embodiment 33 The compound of any one of Embodiments 1 to 13, for use in a method for the treatment of a disease.
  • Embodiment 34 The compound for use of Embodiment 34, wherein the disease is a disease involving fibroblast activation protein (FAP), preferably upregulated expression of fibroblast activation protein (FAP).
  • Embodiment 35 The compound for use of any one of Embodiments 33 to 34, wherein the disease involves cells showing upregulated expression of fibroblast activation protein (FAP), preferably diseased tissue containing cells showing upregulated expression of fibroblast activation protc more preferably disease involving tumor associated fibroblasts.
  • FAP fibroblast activation protein
  • FAP fibroblast activation protein
  • Embodiment 36 The compound for use of any one of Embodiments 33 to 35, wherein the disease is a neoplasm, preferably a cancer or tumor.
  • Embodiment 37 The compound for use of Embodiment 36, wherein the neoplasm, cancer, and tumor are each and individually selected from the group comprising a solid tumor, an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangiocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer, melanoma, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, salivary carcinoma, sarcoma, squamous cell carcinoma, and thyroid cancer.
  • a solid tumor an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangiocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer, melanom
  • Embodiment 38 The compound for use of Embodiment 37, wherein the neoplasm, cancer, and tumor are each and individually selected from the group comprising breast cancer, colorectal cancer, cholangiocarcinoma, head and neck cancer, lung cancer, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, and squamous cell carcinoma.
  • Embodiment 39 The compound for use of any one of Embodiments 33 to 35, wherein the disease is selected from the groups comprising inflammatory disease, cardiovascular disease, autoimmune disease, and fibrotic disease.
  • Embodiment 40 The compound for use of Embodiment 39, wherein the disease is an inflammatory disease.
  • Embodiment 41 The compound for use of Embodiment 40, wherein the disease is atherosclerosis, arthritis, or rheumatoid arthritis.
  • Embodiment 42 The compound for use of Embodiment 39, wherein the disease is a cardiovascular disease.
  • Embodiment 43 The compound for use of Embodiment 42, wherein the diseases is a cardiovascular disease involving atherosclerotic plaques.
  • Embodiment 44 The compound for use of Embodiment 43, wherein the diseases is an atherosclerotic pathology caused by rupture of plaques, acute coronary syndrome, myocardial infarction, thrombosis, or vessel occlusion.
  • Embodiment 45 The compound for use of Embodiment 39, wherein the disease is a fibrotic disease.
  • Embodiment 46 The compound for use of Embodiment 45, wherein the disease is selected form the group comprising idiopathic pulmonary fibrosis, Crohn’s disease, and liver fibrosis.
  • Embodiment 47 The compound for use of any one of Embodiments 33 to 38, wherein the compound comprises a therapeutically active nuclide, preferably a therapeutically active radionuclide.
  • Embodiment 48 The compound for use of Embodiment 47, wherein the therapeutically active nuclide is selected from the group comprising 47 Sc, 67 Cu, 89 Sr, 90 Y, 153 Sm, I49 Tb, !61 Tb, 177 LU, 186 Re, Re, 212 Pb, 213 Bi, 223 Ra, 225 Ac, 22 Tfa, 227 Th, 131 I, 211 At, preferably 47 Sc, 67 Cu,
  • Embodiment 49 The compound for use of any one of Embodiments 33 to 48, wherein the method comprises the administration of a therapeutically effective amount of the compound to a subject, preferably to a mammal, wherein the mammal is selected from the group comprising man, companion animals, pets, and livestock, more preferably the subject is selected from the group comprising man, dog, cat, horse, and cow, and most preferably the subject is a human being.
  • a mammal is selected from the group comprising man, companion animals, pets, and livestock, more preferably the subject is selected from the group comprising man, dog, cat, horse, and cow, and most preferably the subject is a human being.
  • Embodiment 50 Embodiment 50.
  • Embodiment 51 The compound of any one of Embodiments 1 to 13, for use in a method for the selection of a subject from a group of subjects, wherein the subject is likely to respond or likely not to respond to a treatment of a disease, wherein the method for the selection of a subject from a group of subjects comprises carrying out a method of diagnosis using the compound of any one of Embodiments 1 to 13, preferably a method for the diagnosis of a disease as described in any one of Embodiments 14 to 32.
  • Embodiment 52 The compound of any one of Embodiments 1 to 13, for use in a method for the stratification of a group of subjects into subjects which are likely to respond to a treatment of a disease, and into subjects which are not likely to respond to a treatment of a disease, wherein the method for the stratification of a group of subjects comprises carrying out a method of diagnosis using the compound of any one of Embodiments 1 to 13, preferably a method for the diagnosis of a disease as described in any one of Embodiments 14 to 32.
  • Embodiment 53 The compound for use of any one of Embodiments 50 to 52, wherein the disease is a disease involving fibroblast activation protein (FAP), preferably upregulated expression of fibroblast activation protein (FAP).
  • FAP fibroblast activation protein
  • FAP upregulated expression of fibroblast activation protein
  • Embodiment 54 The compound for use of any one of Embodiments 50 to 53, wherein the disease involves cells showing upregulated expression of fibroblast activation protein (FAP), preferably diseased tissue containing cells showing upregulated expression of fibroblast activation protein (FAP), more preferably disease involving tumor associated fibroblasts.
  • Embodiment 55 The compound for use of any one of Embodiments 50 to 54, wherein the disease is a neoplasm, preferably a cancer or tumor.
  • Embodiment 56 The compound for use of Embodiment 55, wherein the neoplasm, cancer, and tumor are each and individually selected from the group comprising a solid tumor, an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangiocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer, melanoma, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, salivary carcinoma, sarcoma, squamous cell carcinoma, and thyroid cancer.
  • a solid tumor an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangiocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer, melanom
  • Embodiment 57 The compound for use of Embodiment 56, wherein the neoplasm, cancer, and tumor are each and individually selected from the group comprising breast cancer, colorectal cancer, cholangiocarcinoma, head and neck cancer, lung cancer, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, and squamous cell carcinoma.
  • Embodiment 58 The compound for use of any one of Embodiments 50 to 54, wherein the disease is selected from the groups comprising inflammatory disease, cardiovascular disease, autoimmune disease, and fibrotic disease.
  • Embodiment 59 The compound for use of Embodiment 58, wherein the disease is an inflammatory disease.
  • Embodiment 60 The compound for use of Embodiment 59, wherein the disease is atherosclerosis, arthritis or rheumatoid arthritis.
  • Embodiment 61 The compound for use of Embodiment 58, wherein the disease is a cardiovascular disease.
  • Embodiment 62. The compound for use of Embodiment 61, wherein the disease is a cardiovascular disease involving atherosclerotic plaques.
  • Embodiment 63 The compound for use of Embodiment 62, wherein the disease is an atherosclerotic pathology caused by rapture of plaques, acute coronary syndrome, myocardial infarction, thrombosis, or vessel occlusion.
  • Embodiment 64 The compound for use of Embodiment 58, wherein the disease is a fibrotic disease.
  • Embodiment 65 The compound for use of Embodiment 64, wherein the disease is selected from the group comprising idiopathic pulmonary fibrosis, Crohn’s disease, and liver fibrosis.
  • Embodiment 66 The compound for use of any one of Embodiments 50 to 65, wherein the method of diagnosis is an imaging method.
  • Embodiment 67 The compound for use of Embodiment 66, wherein the imaging method is selected from the group comprising scintigraphy, Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET).
  • the imaging method is selected from the group comprising scintigraphy, Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET).
  • SPECT Single Photon Emission Computed Tomography
  • PET Positron Emission Tomography
  • Embodiment 68 The compound for use of any one of Embodiments 50 to 67, wherein the compound comprises a diagnostically active nuclide, preferably a diagnostically active radionuclide.
  • Embodiment 69 The compound for use of Embodiment 68, wherein the diagnostically active nuclide is selected from the group comprising 43 Sc, 44 Sc, 51 Mn, 52 MII, 64 Cu, 67 Ga, 6S Ga, 86 Y, S9 Zr, ⁇ Tc, 99lll Tc, 11 frii, 152 Tb, !
  • FAP fibroblast activation protein
  • FAP human fibroblast activation protein
  • Embodiment 71 The compound for use of Embodiment 70, wherein the effector is selected from the group comprising a diagnostically active nuclide and a therapeutically active nuclide.
  • Embodiment 72 The compound for use of Embodiment 71, wherein the diagnostically active nuclide is a diagnostically active radionuclide.
  • Embodiment 73 The compound for use of Embodiment 72, wherein the diagnostically active radionuclide is selected from the group consisting of 43 Sc, 44 Sc, 51 Mn, 52 MII, 64 Cu, 67 Ga, 68 Ga, 86 Y, 89 Zr, 94m Tc, 99m Tc, 11 3 ⁇ 4, 152 Tb, 155 Tb, 20l Tl, 203 Pb, 18 F, 76 Br, 77 Br, 123 1, 124 I, 125 I, preferably 43 Sc, 44 Sc, 64 Cu, 67 Ga, 68 Ga, 86 Y, 89 Zr, 99m Tc, 1 H In, 152 Tb, 155 Tb, 203 Pb, 18 P, 76 Br, 77 Br,
  • Embodiment 74 The compound for use of any one of Embodiments 70 to 73, wherein the fibroblast activation protein expressed by a cell, preferably a fibroblast, a mesenchymal stem cell, smooth muscle cell, a cell of epithelial origin, or an endothelial cell, more preferably a human fibroblast, mesenchymal stem cell, smooth muscle cell, cell of epithelial origin, or endothelial cell, most preferably a human fibroblast, mesenchymal stem cell, smooth muscle cell, cell of epithelial origin, or endothelial cell each showing upregulated expression of fibroblast activation protein (FAP).
  • FAP fibroblast activation protein
  • Embodiment 75 The compound for use of Embodiment 74, wherein the cell is contained in or part of a tissue, preferably a diseased tissue of a subject suffering from a disease.
  • Embodiment 76 The compound for use of Embodiment 75, wherein the disease involves cells showing upregulated expression of fibroblast activation protei; ), preferably diseased tissue containing cells showing upregulated expression of fibroblast activation protein (FAP), more preferably disease involving tumor associated fibroblasts.
  • fibroblast activation protei preferably diseased tissue containing cells showing upregulated expression of fibroblast activation protein (FAP), more preferably disease involving tumor associated fibroblasts.
  • FAP fibroblast activation protein
  • Embodiment 77 The compound for use of any one of Embodiments 75 to 76, wherein the disease is a neoplasm, preferably a cancer or tumor.
  • Embodiment 78 The compound for use of Embodiment 77, wherein the neoplasm, cancer, and tumor are each and individually selected from the group comprising a solid tumor, an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangiocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer, melanoma, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, salivary carcinoma, sarcoma, squamous cell carcinoma, and thyroid cancer.
  • a solid tumor an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangiocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer, mel
  • Embodiment 79 The compound for use of Embodiment 78, wherein the neoplasm, cancer, and tumor are each and individually selected from the group comprising breast cancer, colorectal cancer, cholangiocarcinoma, head and neck cancer, lung cancer, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, and squamous cell carcinoma.
  • Embodiment 80 The compound for use of any one of Embodiments 75 to 76, wherein the disease is selected from the groups comprising inflammatory disease, cardiovascular disease, autoimmune disease, and fibrotic disease.
  • Embodiment 81 The compound for use of Embodiment 80, wherein the disease is an inflammatory disease.
  • Embodiment 82 The compound for use of Embodiment 81, wherein the disease is atherosclerosis, arthritis or rheumatoid arthritis.
  • Embodiment 83 The compound for use of Embodiment 80, wherein the disease is a cardiovascular disease.
  • Embodiment 84 The compound for use of Embodiment 83, wherein the diseases is a cardiovascular disease involving atherosclerotic plaques.
  • Embodiment 85 The compound for use of Embodiment 84, wherein the disease is an atherosclerotic pathology caused by rupture of plaques, acute coronary syndrome, myocardial infarction, thrombosis, or vessel occlusion.
  • Embodiment 86 The compound for use of Embodiment 80, wherein the disease is a fibrotic disease.
  • Embodiment 87 The compound for use of Embodiment 86, wherein the disease is selected form the group comprising idiopathic pulmonary fibrosis, Crohn’s disease, and liver fibrosis.
  • Embodiment 88 The compound for use of Embodiment 71, wherein the therapeutically active nuclide is a therapeutically active radionuclide.
  • Embodiment 89 The compound for use of Embodiment 88, wherein the therapeutically active radionuclide is selected from the group consisting of 47 Sc, 67 Cu, 89 Sr, 90 Y, 153 Sm, 149 Tb, 161 Tb, 177 LU, 186 Re, 188 Re, 2l2 Pb, 21 3 ⁇ 4i, 223 Ra, 225 Ac, 226 Tli, 227 Th, U1 I, 21 'At, preferably 47 Sc, 67 Cu, 90 Y, 177 LU, 188 Re, 2I2 Pb, 21 3 ⁇ 4i, 225 Ac, 227 Th, 131 I, 21 ’At and most preferably 90 Y, 177 Lu, 225 Ac, 227 Tli, 131 1 and 211 At
  • Embodiment 90 The compound for use of any one of Embodiment 88 to 89, wherein the fibroblast activation protein (FAP) is expressed by a cell, preferably a fibroblast, a mesenchymal stem cell, smooth muscle cell, a cell of epithelial origin, or an endothelial cell, more preferably a human fibroblast, mesenchymal stem cell, smooth muscle cell, cell of epithelial origin, or endothelial cell, most preferably a human fibroblast, mesenchymal stem cell, smooth muscle cell, cell of epithelial origin, or endothelial cell showing upregulated expression of fibroblast activation prc i.
  • Embodiment 91 The compound for use of Embodiment 90, wherein the cell is contained in or part of a tissue, preferably a diseased tissue of a subject suffering from a disease.
  • Embodiment 92 The compound for use of Embodiment 91, wherein the disease involves cells showing upregulated expression of fibroblast activation prote preferably diseased tissue containing cells showing upregulated expression of fibroblast activation prot more preferably disease involving tumor associated fibroblasts.
  • Embodiment 93 The compound for use of any one of Embodiments 90 to 92, wherein the disease is a neoplasm, preferably a cancer or tumor.
  • Embodiment 94 The compound for use of Embodiment 93 , wherein the neoplasm, cancer, and tumor are each and individually selected from the group comprising a solid tumor, an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangiocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer, melanoma, mesothelioma, neuroendocrine tumors and carcinomas, ovarian cancer, pancreatic cancer, prostate cancer, renal cell carcinoma, salivary carcinoma, sarcoma, squamous cell carcinoma, and thyroid cancer.
  • a solid tumor an epithelial tumor, bladder cancer, breast cancer, cervical cancer, colorectal cancer, cholangiocarcinoma, endometrial cancer, esophageal cancer, gastric cancer, gastrointestinal stromal tumors, head and neck cancer, liver cancer, lung cancer,
  • Embodiment 95 A composition, preferably a pharmaceutical composition, wherein the composition comprises a compound according to any one of Embodiment 1 to 13 and a pharmaceutically acceptable excipient.
  • Embodiment 96 The composition of Embodiment 95 for use in any method as defined in any of the preceding claims.
  • Embodiment 97 A method for the diagnosis of a disease in a subject, wherein the method comprises administering to the subject a diagnostically effective amount of a compound according to any one of Embodiments 1 to 13.
  • Embodiment 99 A method for the treatment of a disease in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of a compound according to any one of Embodiment 1 to 13.
  • Embodiment 100 The method of Embodiment 99, wherein the compound comprises a therapeutically active agent, whereby the agent is preferably a radionuclide.
  • Embodimer The method of any one of Embodiments 97 to 100, wherein the disease is a disease involving fibroblast activation protein (FAP), preferably upregulated expression of fibroblast activation protei
  • FAP fibroblast activation protein
  • Embodiment 102 The method of any one of Embodiments 97 to 101 , wherein the disease involves cells showing upregulated expression of fibroblast activation prot preferably diseased tissue containing cells showing upregulated expression of fibroblast activation protein more preferably disease involving tumor associated fibroblasts.
  • Embodiment 103 The method of any one of Embodiments 97 to 102, wherein the disease is selected from the groups comprising neoplasms, preferably cancers or tumors, and inflammatory disease, cardiovascular disease, autoimmune disease, and fibrotic disease.
  • the disease is selected from the groups comprising neoplasms, preferably cancers or tumors, and inflammatory disease, cardiovascular disease, autoimmune disease, and fibrotic disease.
  • Embodiment 104 A kit comprising a compound according to any one of Embodiments 1 to 13, one or more optional excipient(s) and optionally one or more device(s), whereby the device(s) is/are selected from the group comprising a labeling device, a purification device, a handling device, a radioprotection device, an analytical device or an administration device.
  • Embodiment 105 kit of Embodiment 104 for use in any method as defined in any of the preceding claims. More specifically, the problem underlying the present invention is solved in a first aspect by a compound selected from the group consisting of compound Hex-[Cys(tMeBn(DOTA-AET»-Pro-Pro-Thr-Gln-Phe-Cys]-OH (3BP-3554) of the following formula
  • the problem underlying the present invention is solved in a second aspect by the compound according to the first aspect, including any embodiment thereof, for use in a method for the diagnosis of a disease. More specifically, the problem underlying the present invention is solved in a third aspect by the compound according to the first aspect, including any embodiment thereof, for use in a method for the treatment of a disease.
  • the problem underlying the present invention is solved in a fourth aspect by the compound according to the first aspect, including any embodiment thereof, for use in a method for the identification of a subject, wherein the subject is likely to respond or likely not to respond to a treatment of a disease, wherein the method for the identification of a subject comprises carrying out a method of diagnosis using the compound according to the first aspect including any embodiment thereof.
  • the problem underlying the present invention is solved in a fifth aspect by the compound according to the first aspect, including any embodiment thereof, for use in a method for the selection of a subject from a group of subjects, wherein the subject is likely to respond or likely not to respond to a treatment of a disease, wherein the method for the selection of a subject from a group of subjects comprises carrying out a method of diagnosis using the compound according to the first aspect, including any embodiment thereof.
  • the problem underlying the present invention is solved in a sixth aspect by the compound according to the first aspect, including any embodiment thereof, for use in a method for the stratification of a group of subjects into subjects which are likely to respond to a treatment of a disease, and into subjects which are not likely to respond to a treatment of a disease, wherein the method for the stratification of a group of subjects comprises carrying out a method of diagnosis using the compound according to the first aspect, including any embodiment thereof.
  • composition preferably a pharmaceutical composition
  • the composition comprises a compound according to the first aspect including any embodiment thereof and a pharmaceutically acceptable excipient.
  • the problem underlying the present invention is solved in an eighth aspect by a method for the diagnosis of a disease in a subject, wherein the method comprises administering to the subject a diagnostically effective amount of a compound according to the first aspect, including any embodiment thereof.
  • the problem underlying the present invention is solved in a ninth aspect by a method for the treatment of a disease in a subject, wherein the method comprises administering to the subject a therapeutically effective amount of a compound according to the first aspect including any embodiment thereof.
  • kits comprising a compound according to the first aspect, including any embodiment thereof, one or more optional excipient(s) and optionally one or more device(s), whereby the device(s) is/are selected from the group comprising a labeling device, a purification device, a handling device, a radioprotection device, an analytical device or an administration device.
  • a or the compound of the invention is any compound disclosed herein, including but not limited to any compound described in any of the above embodiments and any of the following embodiments.
  • composition of the invention is any composition disclosed herein, including but not limited to any composition described in any of the above embodiments and any of the following embodiments.
  • a or the kit of the invention is any kit disclosed herein, including but not limited to any kit described in any of the above embodiments and any of the following embodiments.
  • the present invention is based on the surprising finding of the present inventors that the compound of the invention and more specifically the cyclic peptide thereof provides for a highly specific binding of a compound comprising such cyclic peptide to fibroblast activation protein (FAP), since FAP-specific cyclic peptide-based inhibitors with nanomolar affinity have not been described so far.
  • FAP fibroblast activation protein
  • the present inventors have found that the compounds of the invention are surprisingly stable in blood plasma, and are surprisingly useful as imaging agents and efficacious in shrinking tumors.
  • a chelator is a compound which is capable of forming a chelate, whereby a chelate is a compound, preferably a cyclic compound where a metal or a moiety having an electro r a lone pair of electrons participates in the formation of the ring. More preferably, a chef his kind of compound where a single ligand occupies more than one coordination site at a central atom.
  • a diagnostically active compound is a compound which is suitable for or useful in the diagnosis of a disease.
  • a diagnostic agent or a diagnostically active agent is a compound which is suitable for or useful in the diagnosis of a disease.
  • a therapeutically active compound is a compound which is suitable for or useful in the treatment of a disease.
  • a therapeutic agent or a therapeutically active agent is a compound which is suitable for or useful in the treatment of a disease.
  • a theragnostically active compound is a compound which is suitable for or useful in both the diagnosis and therapy of a disease.
  • a theragnostic agent or a theragnostically active agent is a compound which is suitable for or useful in both the diagnosis and therapy of a disease.
  • theragonstics is a method for the combined diagnosis and therapy of a disease; preferably, the combined diagnostically and therapeutically active compounds used in theragnostics are radiolabeled.
  • treatment of a disease is treatment and/or prevention of a disease.
  • a disease involving is a disease where cells including but not limited to fibroblasts expressing, preferably in an upregulated manner, FAP and tissue either expressing or containing or comprising cells such as fibroblasts, preferably expressing FAP in an upregulated manner respectively, are either a or the cause for the disease and/or the symptoms of the disease, or are part of the pathology underlying the disease.
  • a preferred FAP-expressing cell is a cancer associated fibroblast (CAF).
  • the disease preferably when used in connection with the treatment, treating and/or therapy of the disease, affecting the cells, the tissue and pathology, respectively, results in cure, treatment or amelioration of the disease and/or the symptoms of the disease.
  • labeling of th expressing cells and/or of the FAP-expressing tissue allows discriminating or distinguishing said cells and/or said tissue from healthy o non-expressing cells and/or healthy or FAP non-expressing tissue. More preferably such discrimination or distinction forms the basis for said diagnosis and diagnosing, respectively.
  • labeling means the interaction of a detectable label either directly or indirectly with the FAP-expressing cells and/or with tli> 1 , P ⁇ pressing tissue or tissue containing s xpressing cells; more preferably such interaction involves or is based on the interaction of the label or a compound bearing such label with FAP.
  • a target cell is a cell which is expressin and is a or the cause for a disease and/or the symptoms of a disease, or is part of the pathology underlying a disease.
  • a non-target cell is a cell which is either not express!] is not a or the cause for a disease and/or the symptoms of a disease, or is part of the pathology underlying a disease.
  • a neoplasm is an abnormal new growth of cells.
  • the cells in a neoplasm grow more rapidly than normal cells and will continue to grow if not treated.
  • a neoplasm may be benign or malignant.
  • a tumor is a mass lesion that may be benign or malignant.
  • a cancer is a malignant neoplasm.
  • amino acid sequences of the peptides provided herein are depicted in typical peptide sequence format, as would be understood by the ordinary skilled artisan.
  • the three- letter code of a conventional amino acid, or the code for a non-conventional amino acid or the abbreviations for additional building blocks indicates the presence of the amino acid or building block in a specified position within the peptide sequence.
  • the code for each amino acid or building block is connected to the code for the next and/or previous amino acid or building block in the sequence by a hyphen which (typically represents an amide linkage).
  • amino acid contains more than one amino and/or carboxy group all orientations of this amino acid are in principle possible, but in a-amino acid the utilization of the a-amino and the a-carboxy group is preferred and otherwise preferred orientations are explicitly specified.
  • the first letter indicates the stereochemistry of the C-a- atom if applicable.
  • a capital first letter indicates that the L-form of the amino acid is present in the peptide sequence, while a lower case first letter indicating that the D-form of the correspondent amino acid is present in the peptide sequence.
  • an aromatic L-a-amino acid is any kind of L- ot-amino acid which comprises an aryl group.
  • a heteroaromatic L-a-amino acid is any kind of L-a-amino acid which comprises a heteroaryl group.
  • amino acid sequences are presented herein in N- to C- terminus direction.
  • Compounds of the invention typically contain amino acid sequences as provided herein.
  • Conventional amino acids also referred to as natural amino acids are identified according to their standard three-letter abbreviations and one-letter abbreviations, as set forth in Table 2.
  • Non-conventional amino acids also referred to as non-natural amino acids, are any kind ofnon- oligomeric compound which comprises an amino group and a carboxylic group and is not a conventional amino acid.
  • non-conventional amino acids and other building blocks as used for the construction of compounds of the invention are identified according to their abbreviation or name found in Table 3.
  • the structures of some building blocks are depicted with an exemplary reagent for introducing the building block into the peptide (e.g., as carboxylic acid like) or these building blocks are shown as residue which is completely attached to another structure like a peptide or amino acid.
  • the structures of the amino acids are shown as explicit amino acids and not as residues of the amino acids how they are presented after implementation in the peptide sequence. Some larger chemical moieties consisting of more than one moiety are also shown for the reason of clarity.
  • Table 3 Abbreviation, name and structure of non-natural amino-acid and other building blocks and chemical moieties
  • DOT A stands for 1 ,4,7,10-tetrazacyclododecane- 1 ,4,7, 10-tetraacetic acid.
  • a chelator in the compound of the invention includes, if not stated otherwise, the possibility that the chelator is complexed to any metal complex partner, i.e. any metal which, in principle, can be complexed by the chelator.
  • An explicitly mentioned chelator of a compound of the invention or the general term chelator in connection with the compound of the invention refers either to the uncomplexed chelator as such or to the chelator to which any metal complex partner is bound, wherein the metal complex partner is any radioactive or non-radioactive metal complex partner.
  • the chelator metal complex i.e. the chelator to which the metal complex partner is bound, is a stable chelator metal complex.
  • Non-radioactive chelator metal complexes have several applications, e.g. for assessing properties like stability or activity which are otherwise difficult to determine.
  • cold variants of the radioactive versions of the metal complex partner e.g. non-radioactive Gallium, Lutetium or Indium complexes as described in the examples
  • they are valuable tools for identifying metabolites in vitro or in vivo, as well as for assessing toxicity properties of the compounds of invention.
  • chelator metal complexes can be used in binding assays utilizing the fluorescence properties of some metal complexes with distinct ligands (e.g. Europium salts).
  • the radioactive nuclide which is or which is to be attached to the compound of the invention is selected taking into consideration the disease to be treated and/or the disease to be diagnosed, respectively, and/or the particularities of the patient and patient group, respectively, to be treated and to be diagnosed, respectively.
  • the radioactive nuclide is also referred to as radionuclide.
  • Radioactive decay is the process by which an atomic nucleus of an unstable atom loses energy by emitting ionizing particles (ionizing radiation). There are different types of radioactive decay.
  • the parent and daughter are different chemical elements, and thus the decay process results in nuclear transmutation (creation of an atom of a new element).
  • the radioactive decay can be alpha decay, beta decay, and gamma decay. Alpha decay occurs when the nucleus ejects an alpha particle (helium nucleus).
  • Beta decay occurs when the nucleus emits an electron (b -decay) or positron (P + -decay) and a type of neutrino, in a process that changes a proton to a neutron or the other way around.
  • radioactive decay processes that do not result in transmutation.
  • the energy of an excited nucleus may be emitted as a gamma ray in gamma decay, or used to eject an orbital electron by interaction with the excited nucleus in a process called internal conversion, or used to absorb an inner atomic electron from the electron shell whereby the change of a nuclear proton to neutron causes the emission of an electron neutrino in a process called electron capture (EC), or may be emitted without changing its number of proton and neutrons in a process called isomeric transition (IT).
  • EC electron capture
  • I isomeric transition
  • Another form of radioactive decay, the spontaneous fission (SF) is found only in very heavy chemical elements resulting in a spontaneous breakdown into smaller nuclei and a few isolated nuclear particles.
  • the radionuclide can be used for labeling of the compound of the invention.
  • the radionuclide is suitable for complexing with a chelator, leading to a radionuclide chelate complex.
  • one or more atoms of the compound of the invention are of non-natural isotopic composition, preferably these atoms are radionuclides; more preferably radionuclides of carbon, oxygen, nitrogen, sulfur, phosphorus and halogens: These radioactive atoms are typically part of amino acids, in some case halogen containing amino acids, and/or building blocks and in some cases halogenated building blocks each of the compound of the invention.
  • the radionuclide has a half-life that allows for diagnostic and/or therapeutic medical use. Specifically, the half-life is between 1 min and 100 days.
  • the radionuclide has a decay energy that allows for diagnostic and/or therapeutic medical use.
  • the decay energy is between 0.004 and 10 MeV, preferably between 0.05 and 4 MeV, for diagnostic use.
  • the decay energy is between 0.6 and 13.2 MeV, preferably between 1 and 6 MeV, for diagnostic use.
  • the decay energy is between 0.039 and 10 MeV, preferably between 0.4 and 6.5 MeV, for therapeutic use.
  • the radionuclide is industrially produced for medical use. Specifically, the radionuclide is available in GMP quality.
  • the daughter nuclide(s) after radioactive decay of the radionuclide are compatible with the diagnostic and/or therapeutic medical use. Furthermore, the daughter nuclides are either stable or further decay in a way that does not interfere with or even support the diagnostic and/or therapeutic medical use. Representative radionuclides which may be used in connection with the present invention are summarized in Table 4.
  • the radionuclide is used for diagnosis.
  • the radioactive isotope is selected from the group, but not limited to, comprising 43 Sc, 44 Sc, 5 1 Mu, 52 MJJ, 64 CU, 67 Ga, 68 Ga, 86 Y, 89 Zr, 94m Tc, 99n, Tc, m In, 152 Tb, 155 Tb, m Lu, 201 T1, 203 Pb, 18 P, 76 Br, 77 Br, 123 I, 124 I, 125 I.
  • the radionuclide is selected from the group comprising 43 Sc, 44 Sc, 64 Cu, 67 Ga, 68 Ga, 86 Y, 89 Zr, 99m Tc, l u ln, 152 Tb, 155 Tb, 203 Pb, 18 F, 76 Br, 77 Br, 1 23 I, 124 1, 125 I. Even more preferably, the radionuclide is selected from the group comprising 64 Cu, 68 Ga, 89 Zr, 99m Tc, i n In, 18 F, 123 I, and 124 I. It will however, also be acknowledged by a person skilled in the art that the use of said radionuclide is not limited to diagnostic purposes, but encompasses their use in therapy and tlieragnostics when conjugated to the compound of the invention.
  • the radionuclide is used for therapy.
  • the radioactive isotope is selected from the group comprising 47 Sc, 67 Cu, 89 Sr, 90 Y,“‘to, 153 Sm, 149 Tb, I61 Tb, 177 LU, 186 Re, 188 Re, 212 Pb, 213 Bi, 223 Ra, 225 Ac, 226 Th, 227 Th, ,31 I, 21 ‘At. More preferably, the radioactive isotope is selected from the group comprising 47 Sc, 67 Cu, 90 Y, 177 Lu, 188 Re, 212 Pb, 213 Bi, 225 Ac, 227 Th, 131 1, 211 At.
  • the radionuclide is selected from the group comprising 90 Y, 177 Lu, 225 Ac, 227 Th, 131 I and 211 At. It will however, also be acknowledged by a person skilled in the art that the use of said radionuclide is not limited to therapeutic purposes, but encompasses their use in diagnostic and theragnostics when conjugated to the compound of the invention.
  • the compound of the invention is present as a pharmaceutically acceptable salt.
  • a "pharmaceutically acceptable salt” of the compound of the present invention is preferably an acid salt or a base salt that is generally considered in the art to be suitable for use in contact with the tissues of human beings or animals without excessive toxicity or carcinogenicity, and preferably without irritation, allergic response, or other problem or complication.
  • Such salts include mineral and organic acid salts of basic residues such as amines, as well as alkali or organic salts of acidic residues such as carboxylic acids.
  • Compounds of the invention are capable of forming internal salts which are also pharmaceutically acceptable salts.
  • Suitable pharmaceutically acceptable salts include, but are not limited to, salts of acids such as hydrochloric, phosphoric, hydrobromic, malic, glycolic, fumaric, sulfuric, sulfamic, sulfanilic, formic, toluenesulfonic, methanesulfonic, benzene sulfonic, ethane disulfonic, 2- hydroxyethylsulfonic, nitric, benzoic, 2-acetoxybenzoic, citric, tartaric, lactic, stearic, salicylic, glutamic, ascorbic, pamoic, succinic, fumaric, maleic, propionic, hydroxymaleic, hydroiodic, phenylacetic, alkanoic such as acetic, HOOC-(CH2) n -COOH where n is any integer from 0 to 4, i.e., 0, 1, 2, 3, or 4, and the like.
  • acids such as hydrochloric, phospho
  • pharmaceuticall y acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium and ammonium.
  • a pharmaceutically acceptable acid or base salt can be synthesized from a parent compound that contains a basic or acidic moiety by any conventional chemical method. Briefly, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two.
  • non-aqueous media such as ether, ethyl acetate, ethanol, isopropanol or acetonitrile, is preferred.
  • a "pharmaceutically acceptable solvate" of the compound of the invention is preferably a solvate of the compound of the invention formed by association of one or more solvent molecules to one or more molecules of a compound of the invention.
  • the solvent is one which is generally considered in the art to be suitable for use in contact with the tissues of human beings or animals without excessive toxicity or carcinogenicity, and preferably without irritation, allergic response, or other problem or complication.
  • Such solvent includes an organic solvent such as alcohols, ethers, esters and amines.
  • A“hydrate” of the compound of the invention is formed by association of one or more water molecules to one or more molecules of a compound of the invention.
  • Such hydrate includes but is not limited to a hemi-hydrate, mono-hydrate, dihydrate, trihydrate and tetrahydrate. Independent of the hydrate composition all hydrates are generally considered as pharmaceutically acceptable.
  • the compound of the invention has a high binding affinity to FAP and a high inhibitory activity on FAP. Because of this high binding affinity, the compound of the invention is effective as, useful as and/or suitable as a targeting agent and, if conjugated to another moiety, as a targeting moiety.
  • a targeting agent is an agent which interacts with the target molecule which is in the instant case said FAP. In terms of cells and tissues thus targeted by the compound of the invention any cell and tissue, respectively, expressing said FAP is or may be targeted.
  • the compound interacts with a fibroblast activation protein (FAP), preferably with human having an amino acid sequence of SEQ ID NO: 1 or a homolog thereof, wherein the amino acid sequence of the homolog has an identity of FAP that is at least 85% to the amino acid sequence of SEQ ID NO: 1.
  • FAP fibroblast activation protein
  • the identity is 90%, preferably 95 %, 96 %, 97 %, 98 % or 99%.
  • the identity between two nucleic acid molecules can be determined as known to the person skilled in the art. More specifically, a sequence comparison algorithm may be used for calculating the percent sequence homology for the test sequence(s) relative to the reference sequence, based on the designated program parameters.
  • the test sequence is preferably the sequence or protein or polypeptide which is said to be identical or to be tested whether it is identical, and if so, to what extent, to a different protein or polypeptide, whereby such different protein or polypepetide is also referred to as the reference sequence and is preferably the protein or polypeptide of wild type, more preferably the human FAP of SI NO: 1.
  • Optimal alignment of sequences for comparison can be conducted, e.g., by the local homology algorithm of Smith & Waterman (Smith, et al, Advances in Applied Mathematics, 1981, 2: 482), by the homology alignment algorithm of Needleiiian & Wunsch (Needleman, et al, J Mol Biol, 1970, 48: 443), by the search for similarity method of Pearson & Lipman (Pearson, et al, Proc Natl Acad Sci U S A, 1988, 85: 2444), by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by visual inspection.
  • BLAST basic local alignment search tool
  • NCBI National Center for Biotechnology Information
  • the compound of the invention is used or is for use in a method for the treatment of a disease as disclosed herein.
  • Such method preferably, comprises the step of administering to a subject in need thereof a therapeutically effective amount of the compound of the invention.
  • Such method includes, but is not limited to, curative or adjuvant cancer treatment. It is used as palliative treatment where cure is not possible and the aim is for local disease control or symptomatic relief or as therapeutic treatment where the therapy has survival benefit and it can be curative.
  • the method for the treatment of a disease as disclosed herein includes the treatment of the disease disclosed herein, including tumors and cancer, and may be used either as the primary therapy or as second, third, fourth or last line therapy. It is also within the present invention to combine the compound of the invention with further therapeutic approaches. It is well known to the person skilled in the art that the precise treatment intent including curative, adjuvant, neoadjuvant, therapeutic, or palliative treatment intent will depend on the tumor type, location, and stage, as well as the general health of the patient.
  • the disease is selected from the group comprising neoplasm nos, neoplasm benign, neoplasm uncertain whether benign or malignant, neoplasm malignant, neoplasm metastatic, neoplasm malignant uncertain whether primary or metastatic, tumor cells benign, tumor cells uncertain whether benign or malignant, tumor cells malignant, malignant tumor small cell type, malignant tumor giant cell type, malignant tumor fusiform cell type, epithelial neoplasms nos, epithelial tumor benign, carcinoma in situ nos, carcinoma nos, carcinoma metastatic nos, carcinomatosis, epithelioma benign, epithelioma malignant, large cell carcinoma nos, carcinoma undifferentiated type nos, carcinoma anaplastic type nos, pleomorphic carcinoma, giant cell and spindle cell carcinoma, giant cell carcinoma, spindle cell carcinoma, pseudosarcomatous carcinoma, polygonal cell carcinoma, spheroidal cell carcinoma, tumorlet, small cell carcinoma nos, oat cell carcinoma, small cell carcinoma, fu
  • the disease is selected from the group comprising tumors of pancreas, pancreatic adenocarcinoma, tumors of head of pancreas, of body of pancreas, of tail of pancreas, of pancreatic duct, of islets oflangerhans, neck of pancreas, tumor of prostate, prostate adenocarcinoma, prostate gland, neuroendocrine tumors, breast cancer, tumor of central portion ofbreast, upper inner quadrant of breast, lower inner quadrant ofbreast, upper outer quadrant of breast, lower outer quadrant of breast, axillary tail of breast, overlapping lesion of breast, juvenile carcinoma of the breast, tumors of parathyroid gland, myeloma, lung cancer, small cell lung cancer, non-small cell lung cancer, tumor of main bronchus, of upper lobe lung, of middle lobe lung, of lower lobe lung, colorectal carcinoma, tumor of ascending colon, of hepatic flexure of
  • the aforementioned indications may occur in organs and tissues selected from the group comprising external upper lip, external lower lip, external lip nos, upper lip mucosa, lower lip mucosa, mucosa lip nos, commissure lip, overlapping lesion of lip, base of tongue nos, dorsal surface tongue nos, border of tongue, ventral surface of tongue nos, anterior 2/3 of tongue nos, lingual tonsil, overlapping lesion of tongue, tongue nos, upper gum, lower gum, gum nos, anterior floor of mouth, lateral floor of mouth, overlapping lesion of floor of mouth, floor of mouth nos, hard palate, soft palate nos, uvula, overlapping lesion of palate, palate nos, cheek mucosa, vestibule of mouth, retromolar area, overlapping lesion of other and unspecified parts of mouth, mouth nos, parotid gland, submaxillary gland, sublingual gland, overlapping lesion of major salivary glands, major salivary gland nos, tonsillar fossa, tonsillar pillar,
  • the subjects treated with the presently disclosed and claimed compounds may be treated in combination with other non-surgical anti-proliferative (e.g., anti-cancer) drug therapy.
  • the compounds may be administered in combination with an anti-cancer compound such as a cytostatic compound.
  • a cytostatic compound is a compound (e.g., a small molecule, a nucleic acid, or a protein) that suppresses cell growth and/or proliferation.
  • the cytostatic compound is directed towards the malignant cells of a tumor.
  • the cytostatic compound is one which inhibits the growth and/or proliferation of vascular smooth muscle cells or fibroblasts.
  • Suitable anti-proliferative drugs or cytostatic compounds to be used in combination with the presently disclosed and claimed compounds include anti-cancer drags.
  • Numerous anti-cancer drugs which may be used are well known and include, but are not limited to: Acivicin; Aclarubicin; Acodazole Hydrochloride; Acronine; Adozelesin; Aldesleukin; Altretamine; Ambomycin; Ametantrone Acetate; Aminoglutethimide; Amsacrine; Anastrozole; Anthramycin; Asparaginase; Asperlin; Azacitidine; Azetepa; Azotomycin; Batimastat; Benzodepa; Bicalutamide; Bisantrene Hydrochloride; Bisnafide Dimesylate; Bizelesin; Bleomycin Sulfate; Brequinar Sodium; Bropirimine; Busulfan; Cactinomycin; Calusterone; Caracemide; Carbetimer;
  • Temoporfin Teniposide; Teroxirone; Testolactone; Thiamiprine; Thioguanine; Thiotepa; Tiazofurin; Tirapazamine; Topotecan Hydrochloride; Toremifene Citrate; Trestolone Acetate; Triciribine Phosphate; Trimetrexate; Trimetrexate Glucuronate; Tubulozole Hydrochloride; Uracil Mustard; Uredepa; Vapreotide; Velaparib; Verteporfin; Vinblastine Sulfate; Vincristine Sulfate; Vindesine; Vindesine Sulfate; Vinepidine Sulfate; Vinglycinate Sulfate; Vinleurosine Sulfate; Vinorelbine Tartrate; Vinrosidine Sulfate; Vinzolidine Sulfate; Vorozole; Zeniplatin; Zinostatin; and Zorubicin Hydrochloride.
  • anti-cancer drugs include, but are not limited to: 20-epi-l,25 dihydroxyvitamin D3; 5- ethynyluracil; abiraterone; acylfulvene; adecypenol; adozelesin; ALL-TK antagonists; ambamustine; amidox; amifostine; aminolevulinic acid; amrubicin; anagrelide; andrographolide; angiogenesis inhibitors; antagonist D; antagonist G; antarelix; anti- dorsalizing morphogenetic protein- 1 ; antiestrogen; antineoplaston; antisense oligonucleotides; aphidicolin glycinate; apoptosis gene modulators; apoptosis regulators; apurinic acid; ara-CDP- DL-PTBA; arginine deaminase; asulacrine; atamestane; atrimustine; axinastat
  • PARP Poly(ADP-ribose) polymerases
  • PARP inhibitors include but are not limited to olaparib, rupacarib, velaparib, niraparib, talazoparib, pamiparib, iniparib, E7449, and A-966492.
  • inhibitors include but are not limited to inhibitors of ATM and ATR kinases, checkpoint kinase 1 and 2, DNA-dependen protein kinase, and WEE1 kinase (Pilie, et al, Nat Rev Clin Oncol, 2019, 16: 81).
  • a Cyclin-D-Kinase Trends Cell Biol 2018, antibody being capable of binding to a tumor cell and/or metastases and being capable of inducing antibody-dependent cellular cytotoxicity (ADCC) (Kellner, et al, Transfus Med Hemother, 24 f 7 :4: 327), a T cell- or NK cell engager (e.g. bispecific antibodir ' 1 1., et al, J Cancer Res Clin Oncol, 2® 19, 145: 941), a cellular therapy using expanded autologous or allogeneic immune cells (e.g.
  • ADCC antibody-dependent cellular cytotoxicity
  • Immune checkpoint inhibitors induce but are not limited to nivolumab, ipilimumab, pembrolizumab, atezolizumab, avelumab, durvalumab, and cemiplimab.
  • the compounds may be administered prior to, concurrent with, or following other anti-cancer compounds.
  • the administration schedule may involve administering the different agents in an alternating fashion.
  • the compounds may be delivered before and during, or during and after, or before and after treatment with other therapies.
  • the compound is administered more than 24 hours before the administration of the other anti-proliferative treatment.
  • more than one anti-proliferative therapy may be administered to a subject.
  • the subject may receive the present compounds, in combination with both surgery and at least one other anti-proliferative compound.
  • the compound may be administered in combination with more than one anti-cancer drug.
  • the compounds of the present invention are used to detect cells and tissues overexpressi whereby such detection is achieved by conjugating a detectable label to the compounds of the invention, preferably a detectable radionuclide.
  • the cells and tissues detected are diseased cells and tissues and/or are either a or the cause for the disease and/or the symptoms of the disease, or are part of the pathology underlying the disease.
  • the diseased cells and tissues are causing and/or are part of an oncology indicati neoplasms, tumors, and cancers) or a non-oncology indication (e.g. inflammatory disease, cardiovascular disease, autoimmune disease, and fibrotic disease).
  • the compounds of the present invention are used to treat cells and tissues overexpressing preferred embodiment, the cells and tissues treated are diseased cells and tissues and/or are either a or the cause for the disease and/or the symptoms of the disease, or so: art of the pathology underlying the disease.
  • the diseased cells and tissues are causing and/or are part of an oncology indication (e.g. neoplasms, tumors, and cancers) and the therapeutic activity is achieved by conjugating therapeutically active effector to the compounds of the present invention, preferably a therapeutically active radionuclide.
  • the diseased cells and tissues are causing and/or are part of a non-oncology indication (e.g. inflammatory disease, cardiovascular disease, autoimmune disease, and fibrotic disease) and the therapeutic activity is achieved by inhibition of the enzymatic activity of FAP.
  • the compounds of the present invention are administered in therapeutically effective amounts; preferably the compound of the present invention does not comprise a therapeutically active nuclide.
  • An effective amount is a dosage of the compound sufficient to provide a therapeutically or medically desirable result or effect in the subject to which the compound is administered. The effective amount will vary with the particular condition being treated, the age and physical condition of the subject being treated, the severity of the condition, the duration of the treatment, the nature of the concurrent or combination therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner.
  • an effective amount to inhibit proliferation would be an amount sufficient to reduce or halt altogether the abnormal cell proliferation so as to slow or halt the development of or the progression of a cell mass such as, for example, a tumor.
  • “inhibit” embraces all of the foregoing.
  • a therapeutically effective amount will be an amount necessary to extend the dormancy of micrometastases or to stabilize any residual primary tumor cells following surgical or drug therapy.
  • a therapeutically effective amount when using an unconjugated compound without a therapeutically active radionuclide, a therapeutically effective amount will vary with the subject's age, condition, and sex, as well as the nature and extent of the disease in the subject, all of which can be determined by one of ordinary skill in the art.
  • the dosage may be adjusted by the individual physician or veterinarian, particularly in the event of any complication.
  • a therapeutically effective amount is typically, but not limited to, an amount in a range from 0.1 pg/kg to about 2000 mg/kg, or from 1.0 pg/kg to about 1000 mg/kg, or from about 0.1 mg/kg to about 500 mg/kg, or from about 1.0 mg/kg to about 100 mg/kg, in one or more dose administrations daily, for one or more days.
  • the effective daily dose of the active compound may be administered as two, three, four, five, six, or more sub-doses for example administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms.
  • the compounds are administered for more than 7 days, more than 10 days, more than 14 days and more than 20 days.
  • the compound is administered over a period of weeks, or months.
  • the compound is delivered on alternate days. For example, the agent is delivered every two days, or every three days, or every four days, or every five days, or every six days, or every week, or every month.
  • the compound of the present invention is for use in the treatment and/or prevention of a disease, whereby such treatment is radionuclide therapy.
  • radionuclide therapy makes use of or is based on different forms of radiation emitted by a radionuclide.
  • radiation can, for example, be any one of radiation of photons, radiation of electrons including but not limited to b -particles and Auger-electrons, radiation of protons, radiation of neutrons, radiation of positrons, radiation of a-particles or an ion beam.
  • radionuclide therapy can, for example, be distinguished as photon radionuclide therapy, electron radionuclide therapy, proton radionuclide therapy, neutron radionuclide therapy, positron radionuclide therapy, a-particle radionuclide therapy or ion beam radionuclide therapy. All of these forms of radionuclide therapy are encompassed by the present invention, and all of these forms of radionuclide therapy can be realized by the compound of the invention, preferably under the proviso that the radionuclide attached to the compound of the invention, more preferably as an effector, is providing for this kind of radiation.
  • Radionuclide therapy preferably works by damaging the DNA of cells.
  • the damage is caused by a photon, electron, proton, neutron, positron, a-particle or ion ectly or indirectly ionizing the atoms which make up the DNA chain.
  • Indirect ionization happens as a result of the ionization of water, forming free radicals, notably hydroxyl radicals, which then damage the DNA.
  • Oxygen is a potent radiosensitizer, increasing the effectiveness of a given dose of radiation by forming DNA-damaging free radicals. Therefore, use of high pressure oxygen tanks, blood substitutes that carry increased oxygen, hypoxic cell radiosensitizers such as misonidazole and metronidazole, and hypoxic cytotoxins, such as tirapazamine may be applied.
  • Radioactive dose Other factors that are considered when selecting a radioactive dose include whether the patient is receiving chemotherapy, whether radiation therapy is being administered before or after surgery, and the degree of success of surgery.
  • the total radioactive dose may be fractionated, i.e. spread out over time in one or more treatments for several important reasons. Fractionation allows normal cells time to recover, while tumor cells are generally less efficient in repair between fractions. Fractionation also allows tumor cells that were in a relatively radio-resistant phase of the cell cycle during one treatment to cycle into a sensitive phase of the cycle before the next fraction is given. Similarly, tumor cells that were chronically or acutely hypoxic and, therefore, more radioresistant, may reoxygenate between fractions, improving the tumor cell kill.
  • radiosensitivity of a particular tumor which to some extent is a laboratory measure, from "curability" of a cancer by an internally delivered radioactive dose in actual clinical practice.
  • leukemias are not generally curable with radiotherapy, because they are disseminated through the body. Lymphoma may be radically curable if it is localized to one area of the body.
  • many of the common, moderately radioresponsive tumors can be treated with curative doses of radioactivity if they are at an early stage. This applies, for example, to non-melanoma skin cancer, head and neck cancer, non-small cell lung cancer, cervical cancer, anal cancer, prostate cancer.
  • the response of a tumor to radiotherapy is also related to its size. For complex reasons, very large tumors respond less well to radiation than smaller tumors or microscopic disease.
  • Various strategies are used to overcome this effect. The most common technique is surgical resection prior to radiotherapy. This is most commonly seen in the treatment of breast cancer with wide local excision or mastectomy followed by adjuvant radiotherapy. Another method is to shrink the tumor with neoadjuvant chemotherapy prior to radical radionuclide therapy.
  • a third technique is to enhance the radiosensitivity of the cancer by giving certain drugs during a course of radiotherapy. Examples of radiosensiting drugs include, but are not limited to Cisplatin, Nimorazole, and Cetuximab.
  • Introperative radiotherapy is a special type of radiotherapy that is delivered immediately after surgical removal of the cancer. This method has been employed in breast cancer (TARGeted Introperative radioTherapy), brain tumors and rectal cancers.
  • Radionuclide therapy is in itself painless. Many low-dose palliative treatments cause minimal or no side effects. Treatment to higher doses may cause varying side effects during treatment (acute side effects), in the months or years following treatment (long-term side effects), or after re-treatment (cumulative side effects). The nature, severity, and longevity of side effects depends on the organs that receive the radiation, the treatment itself (type of radionuclide, dose, fractionation, concurrent chemotherapy), and the patient.
  • the method for the treatment of a disease of the invention may realize each and any of the above strategies which are as such known in the art, and which insofar constitute further embodiments of the invention.
  • the compound of the invention is used in a method for the diagnosis of a disease as disclosed herein.
  • Such method preferably, comprises the step of administering to a subject in need thereof a diagnostically effective amount of the compound of the invention.
  • an imaging method is selected from the group consisting of scintigraphy, Single Photon Emission Computed Tomography (SPECT) and Positron Emission Tomography (PET).
  • SPECT Single Photon Emission Computed Tomography
  • PET Positron Emission Tomography
  • Scintigraphy is a form of diagnostic test or method used in nuclear medicine, wherein radiopharmaceuticals are internalized by cells, tissues and/or organs, preferably internalized in vivo, and radiation emitted by said internalized radiopharmaceuticals is captured by external detectors (gamma cameras) to form and display two-dimensional images.
  • SPECT and PET forms and displays three-dimensional images. Because of this, SPECT and PET are classified as separate techniques to scintigraphy, although they also use gamma cameras to detect internal radiation. Scintigraphy is unlike a diagnostic X-ray where external radiation is passed through the body to form an image.
  • Single Photon Emission Tomography (SPECT) scans are a type of nuclear imaging technique using gamma rays. They are very similar to conventional nuclear medicine planar imaging using a gamma camera. Before the SPECT scan, the patient is injected with a radiolabeled chemical emitting gamma rays that can be detected by the scanner. A computer collects the information from the gamma camera and translates this into two-dimensional cross-sections. These cross- sections can be added back together to form a three-dimensional image of an organ or a tissue. SPECT involves detection of gamma rays emitted singly, and sequentially, by the radionuclide provided by the radiolabeled chemical.
  • SPECT involves detection of gamma rays emitted singly, and sequentially, by the radionuclide provided by the radiolabeled chemical.
  • the gamma camera is rotated around the patient. Projections are acquired at defined points during the rotation, typically every 3 - 6 degrees, in most cases, a foil 360 degree rotation is used to obtain an optimal reconstruction. The time taken to obtain each projection is also variable, but 15 - 20 seconds is typical. This gives a total scan time of 15 - 20 minutes. Multi-headed gamma cameras are faster.
  • Radiopharmaceuticals Since SPECT acquisition is very similar to planar gamma camera imaging, the same radiopharmaceuticals may be used.
  • PET Positron Emitting Tomography
  • Traditional diagnostic techniques such as X-rays, CT scans, or MRI, produce images of the body's anatomy or structure. The premise with these techniques is that any changes in structure or anatomy associated with a disease can be seen. Biochemical processes are also altered by a disease, and may occur before any gross changes in anatomy. PET is an imaging technique that can visualize some of these early biochemical changes. PET scanners rely on radiation emitted from the patient to create the images.
  • Each patient is given a minute amount of a radioactive pharmaceutical that either closely resembles a natural substance used by the body or binds specifically to a receptor or molecular structure.
  • positron emission decay also known as positive beta decay
  • the radioisotope undergoes positron emission decay (also known as positive beta decay)
  • positron emission decay also known as positive beta decay
  • the positron After traveling up to a few millimeters, the positron encounters an electron and annihilates, producing a pair of annihilation (gamma) photons moving in opposite directions. These are detected when they reach a scintillation material in the scanning device, creating a burst of light, which is detected by photomultiplier tubes or silicon avalanche photodiodes.
  • the technique depends on simultaneous or coincident detection of the pair of photons. Photons that do not arrive in pairs, i.e., within a few nanoseconds, are ignored. All coincidences are forwarded to the image processing unit where the final image data is produced using image reconstruction procedures.
  • SPECT/CT and PET/CT is the combination of SPECT and PET with computed tomography (CT).
  • CT computed tomography
  • the method for the diagnosis of a disease of the invention may realize each and any of the above strategies which are as such known in the art, and which insofar constitute further embodiments of the invention.
  • Compounds of the present invention are useful to stratify patients, i.e. to create subsets within a patient population that provide more detailed information about how the patient will respond to a given drug.
  • Stratification can be a critical component to transforming a clinical trial from a negative or neutral outcome to one with a positive outcome by identifying the subset of the population most likely to respond to a novel therapy.
  • Stratification includes the identification of a group of patients with shared "biological" characteristics to select the optimal management for the patients and achieve the best possible outcome in terms of risk assessment, risk prevention and achievement of the optimal treatment outcome
  • a compound of the present invention may be used to assess or detect, a specific disease as early as possible (which is a diagnostic use), the risk of developing a disease (which is a susceptibility/risk use), the evolution of a disease including indolent vs is a prognostic use) and it may be used to predict the response and the toxicity to a given treatment (which is a predictive use).
  • the compound of the invention is used in a theragnostic method.
  • the concept of theragnostics is to combine a therapeutic agent with a corresponding diagnostic test that can increase the clinical use of the therapeutic drug.
  • the concept of theragnostics is becoming increasingly attractive and is widely considered the key to improving the efficiency of drug treatment by helping doctors identify patients who might profit from a given therapy and hence avoid unnecessary treatments.
  • a compound of the present invention is used for the diagnosis of a patient, i.e. identification and localization of the primary tumor mass as well as potential local and distant metastases. Furthermore, the tumor volume can be determined, especially utilizing three-dimensional diagnostic modalities such as SPECT or PET. Only those patients having SPECT or PET.
  • such therapy is a FAP -targeted therapy using a compound of the present invention.
  • a compound of the present invention chemically identical tumor-targeted diagnostics, preferably imaging diagnostics for scintigraphy, PET or SPECT and radiotherapeutics are applied.
  • Such compounds only differ in the radionuclide and therefore usually have a very similar if not identical pharmacokinetic profile. This can be realized using a chelator and a diagnostic or therapeutic radiometal. Alternatively, this can be realized using a precursor for radiolabeling and radiolabeling with either a diagnostic or a therapeutic radionuclide.
  • diagnostic imaging is used preferably by means of quantification of the radiation of the diagnostic radionuclide and subsequent dosimetry which is known to those skilled in the art and the prediction of drug concentrations in the tumor compared to vulnerable side effect organs.
  • a truly individualized drug dosing therapy for the patient is achieved.
  • the theragnostic method is realized with only one theragnostically active compound such as a compound of the present invention labeled with a radionuclide emitting diagnostically detectable radiation (e.g. positrons or gamma rays) as well as therapeutically effective radiation (e.g. electrons or alpha particles).
  • diagnostically detectable radiation e.g. positrons or gamma rays
  • therapeutically effective radiation e.g. electrons or alpha particles
  • the invention also contemplates a method of intraoperatively identifying/disclosing diseased tissues ibject. Such method uses a compound of the invention, whereby such compound of the invention preferably comprises as Effector a diagnostically active agent.
  • the compound of the invention may Joyed as adjunct or adjuvant to any other tumor treatment including, surgery as the primary method of treatment of most isolated solid cancers, radiation therapy involving the use of ionizing radiation in an attempt to either cure or improve the symptoms of cancer using either sealed internal sources in the form of brachytherapy or external sources, chemotherapy such as alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and other antitumor agents, hormone treatments that modulate tumor cell behavior without directly attacking those cells, targeted agents which directly target a molecular abnormality in certain types of cancer including monoclonal antibodies and tyrosine kinase inhibitors, angiogenesis inhibitors, immunotherapy, cancer vaccination, palliative care including actions to reduce the physical, emotional, spiritual, and psycho-social distress to improve the patient's quality of life and alternative treatments including a diverse group of health care systems, practices, and products that are
  • the subject is a patient.
  • a patient is a subject which has been diagnosed as suffering from or which is suspected of suffering from or which is at risk of suffering from or developing a disease, whereby the disease is a disease as described herein and preferably a disease involving
  • Dosages employed in practicing the methods for treatment and diagnosis, respectively, where a radionuclide is used and more specifically attached to or part of the compound of the invention will vary depending e.g. on the particular condition to be treated, for example the known radiosensitivity of the tumn ; y s, toe volume of the tumor and the therapy desired. In general, the dose is calculated on the basis of radioactivity distribution to each organ and on observed target uptake.
  • a g-emitting complex may be administered once or at several times for diagnostic imaging. In animals, an indicated dose range may be from 0.1 pg/kg to 5 mg/kg of the compound of the invention complexed e.g. with 1 to 200 MBq of In or 89 Zr.
  • a b-emitting complex of the compound of the invention may be administered at several time points e.g. over a period of 1 to 3 weeks or longer.
  • an indicated dosage range may be of from 0.1 pg kg to 5 mg/kg of the compound of the invention complexed e.g. with 1 to 200 °Y or m Lu.
  • an indicated dosage range is from u. i to 100 pg/kg of the compound of the invention complexed with e.g. 10 to 400 MBq In or 89 Zr.
  • an indicated dosage range is of from 0.1 to 100 pg/kg of the compound of the invention complexed with e.g. 10 to 5000 MBq 90 Y or 177 Lu,
  • the instant invention is related to a composition and a pharmaceutical composition in particular, comprising the compound of the invention.
  • the pharmaceutical composition of the present invention comprises at least one compound of the invention and, optionally, one or more carrier substances, excipients and/or adjuvants.
  • the pharmaceutical composition may additionally comprise, for example, one or more of water, buffers such as, e.g., neutral buffered saline or phosphate buffered saline, ethanol, mineral oil, vegetable oil, dimethylsulfoxide, carbohydrates such as e.g., glucose, mannose, sucrose or dextrans, mannitol, proteins, adjuvants, polypeptides or amino acids such as glycine, antioxidants, chelating agents such as EDTA or glutathione and/or preservatives.
  • buffers such as, e.g., neutral buffered saline or phosphate buffered saline
  • ethanol mineral oil
  • vegetable oil dimethylsulfoxide
  • carbohydrates such as e.g., glucose, mannose, sucrose or dextrans, mannitol
  • composition of the invention may be formulated for any appropriate route of administration, including, for example, topical such as, e.g., transdemia1 or ocular, oral, buccal, nasal, vaginal, rectal or parenteral administration.
  • parenteral as used herein includes subcutaneous, intradermal, intravascular such as, e.g., intravenous, intramuscular, intrathecal and intraperitoneal injection, as well as any similar injection or infusion technique.
  • a preferred route of administration is intravenous administration.
  • the compound of the invention comprising a radionuclide is administered by any conventional route, in particular intravenously, e.g. in the form of injectable solutions or suspensions.
  • the compound of the invention may also be administered advantageously by infusion, e.g., by an infusion of 30 to 60 min. on the site of the tumor, the compound of the invention may be administered as close as possible to the tumor site, e.g. by means of a catheter. Such administration may be carried out directly into the tumor tissue or into the surrounding tissue or into the afferent blood vessels.
  • the compound of the invention may also be administered repeatedly in doses, preferably in divided doses.
  • a pharmaceutical composition of the invention comprises a stabilizer, e.g. a free radical scavenger, which inhibits autoradiolysis of the compound of the invention.
  • Suitable stabilizers include, e.g., serum albumin, ascorbic acid, retinol, geiitisic acid or a derivative thereof, or an amino acid infusion solution such, e.g., used for parenteral protein feeding, preferably free from electrolyte and glucose, for example a commercially available amino acid infusion such as Proteinsteril® KE Nephro. Ascorbic acid and gentisic acid are preferred.
  • a pharmaceutical composition of the invention may comprise further additives, e.g.
  • the stabilizer is added to the non-radioactive compound of the invention and introduction of the radionuclide, for instance the complexation with the radionuclide, is performed in the presence of the stabilizer, either at room temperature or, preferably, at a temperature of from 40 to 120° C.
  • the complexation may conveniently be performed under air free conditions, e.g. under N 2 or Ar. Further stabilizer may be added to the composition after complexation.
  • Excretion of the compound of the invention essentially takes place through the kidneys.
  • Further protection of the kidneys from radioactivity accumulation may be achieved by administration of lysine or arginine or an amino acid solution having a high content of lysine and/or arginine, e.g. a commercially available amino acid solution such as Synthamin ® -! 4 or -10, prior to the injection of or together with the compound of the invention, particularly if the Effector is a radionuclide.
  • Protection of the kidneys may also be achieved by administration of plasma expanders such as e.g. gelofusine, either instead of or in addition to amino acid infusion.
  • a pharmaceutical composition of the invention may contain, apart from a compound of the invention, at least one of these further compounds intended for or suitable for kidney protection, preferably kidney protection of the subject to which the compound of the invention is administered.
  • composition of the invention and the pharmaceutical composition of the invention can be equally used in said various methods. It will also be understood by a person skilled in the art that the composition of the invention and the pharmaceutical composition are disclosed herein for use in various methods. It will be equally understood by a person skilled in the art that the compound of the invention can be equally used in said various methods. It will be acknowledged by a person skilled in the art that the composition of the invention and the pharmaceutical composition of the invention contain one or more further compounds in addition to the compound of the invention.
  • Such one or more further compounds can be administered separately from the compound of the invention to the subject which is exposed to or the subject of a method of the invention. Such administration of the one or more further compounds can be performed prior, concurrently with or after the administration of the compound of the invention. It will also be acknowledged by a person skilled in the art that in a method of the invention, apart from a compound of the invention, one or more further compound may be administered to a subject. Such administration of the one or more further compounds can be performed prior, concurrently with or after the administration of the compound of the invention.
  • such one or more further compounds are part of a composition of the invention and/or of a pharmaceutical composition of the invention. It is within the present invention that the compound of the invention and the one or more further compounds may be contained in the same or a different formulation. It is also wi thin the present invention that the compound of the invention and the one or more further compounds are not contained in the same formulation, but are contained in the same package containing a first formulation comprising a compound of the invention, and a second formulation comprising the one or more further compounds, whereby the type of formulation may be the same or may be different.
  • one type of a compound of the invention is contained in the composition of the invention and/or the pharmaceutical composition of the invention. It is also within the present invention that more than one type of a compound of the invention is used, preferably administered, in a method of the invention.
  • composition of the invention and a pharmaceutical composition of the invention may be manufactured in conventional manner.
  • Radiopharmaceutical s have decreasing content of radioactivity with time, as a consequence of the radioactive decay.
  • the physical half-life of the radionuclide is often short for radiopharmaceutical diagnostics. In these cases, the final preparation has to be done shortly before administration to the patient. This is in particular the case for positron emitting radiopharmaceuticals for tomography (PET radiopharmaceuti cals) . It often leads to the use of semi-manufactured products such as radionuclide generators, radioactive precursors and kits.
  • a kit of the invention comprises apart from one or more than one compounds of the invention typically at least one of the followings: instructions for use, final preparation and/or quality control, one or more optional excipient(s), one or more optional reagents for the labeling procedure, optionally one or more radionuclide(s) with or without shielded containers, and optionally one or more device(s), whereby the device(s) is/are selected from the group comprising a labeling device, a purification device, an analytical device, a handling device, a radioprotection device or an administration device.
  • Shielded containers known as "pigs" for general handling and transport of radiopharmaceutical containers come in various configurations for holding radiopharmaceutical containers such as bottles, vials, syringes, etc.
  • One form often includes a removable cover that allows access to the held radiopharmaceutical container. When the pig cover is in place, the radiation exposure is acceptable.
  • a labeling device is selected from the group of open reactors, closed reactors, microfluidic systems, nanoreactors, cartridges, pressure vessels, vials, temperature controllable reactors, mixing or shaking reactors and combinations thereof.
  • a purification device is preferably selected from the group of ion exchange chromatography columns or devices, size-exclusion chromatography columns or devices, affinity chromatography columns or devices, gas or liquid chromatography columns or devices, solid phase extraction columns or devices, filtering devices, centrifugations vials columns or devices.
  • An analytical device is preferably selected from the group of tests or test devices to determine the identity, radiochemical purity, radionuclidic purity, content of radioactivity and specific radioactivity of the radiolabelled compound.
  • a handling device is preferably selected from the group consisting of devices for mixing, diluting, dispensing, labeling, injecting and administering radiopharmaceuticals to a subject.
  • a radioprotection device is used in order to protect doctors and other personnel from radiation when using therapeutic or diagnostic radionuclides.
  • the radioprotection device is preferably selected from the group consisting of devices with protective barriers of radiation-absorbing material selected from the group consisting of aluminum, plastics, wood, lead, iron, lead glass, water, rubber, plastic, cloth, devices ensuring adequate distances from the radiation sources, devices reducing exposure time to the radionuclide, devices restricting inhalation, ingestion, or other modes of entry of radioactive material into the body and devices providing combinations of these measures.
  • An administration device is preferably selected from the group of syringes, shielded syringes, needles, pumps, and infusion devices.
  • Syringe shields are commonly hollow cylindrical structures that accommodate the cylindrical body of the syringe and are constructed of lead or tungsten with a lead glass window that allows the handler to view the syringe plunger and liquid volume within the syringe.
  • Fig. 1 shows a radiochromatogram of 1 r/ Lu-3BP-3407 in formulation buffer containing 100 mg/mL ascorbate and 5 mg/mL fr-niethionine analyzed immediately after synthesis;
  • Fig. 2 shows a radiochromatogram of 1T7 Lii-3BP-3407 in formulation buffer containing 100 mg/mL ascorbate and 5 m; -methionine analyzed six days after synthesis;
  • Fig. 3 shows a radiochromatogram of Lii-3BP-3SS4 in formulation buffer containing 100 mg/mL ascorbate and 5 mg/mL Z-methionine analyzed immediately after synthesis;
  • Fig. 4 shows a radiochromatogram of Lu-3BP-3554 in formulation buffer containing 100 mg/mL ascorbate and 5 mg/mL Z-methionine analyzed six days after synthesis;
  • Fig. 5 shows the percentage of injected dose per gram of tissue (%lD/g) uptake in the kidney, liver, bloodpool, and tumor as determined by SPECT-imaging of“frii-SBP-SdO? lh, 3h, 6h and 24h post injection into the mouse model;
  • Fig. 6 shows the %ID/g uptake in kidney, liver, bloodpool, and HEK-FAP tumor as determined by SPECT-imaging of 1 n Ii i and 24h post injection into the mouse model
  • Fig. 7 shows SPECT-images of l ll In-3BP-3554 1 h, 3 h, 6 h, 24 h and 48 h post injection into mice tumors;
  • Fig. 8 shows the amino acid sequences of human fibroblast activating prof human dipeptidyl peptidase 4 (DDP4) and human prolyl endopeptidase (PREP);
  • Fig. 9 A shows tumor growth over time in mice with HEK-FAP tumors treated with vehicle, cold compound nat Lu-3BP-3554, 30 MBq (low dose) 177 LII ⁇ 3BP ⁇ 3554, and 60 MBq (high dose)
  • Fig. 9 B shows percent body weight changes over time in mice with HEK-FAP tumors treated with vehicle, cold compound nat Lii-3BP-3554, 30 MBq Lu-3BP-3SS4, and 60 MBq m Lu- 3BP-3554;
  • Fig. 10 A shows representative SPECT/CT images over time of the biodistribution of 60 MBq 177 Lu-3BP-3554 in mice with HEK-FAP tumors; ; , nows representative SPECT/CT images over time of the biodistribution of 30 MBq , 77 Lu-3BP-3554 in mice with HEK-FAP tumors;
  • Fig. 11 A shows representative SPECT/CT images of four different sarcoma PDX models 3 h after n i In-3BP ⁇ 3S54 administration;
  • Fig. 11 B shows %ID/g uptake of 1 ! I III-3BP-3554 in four different sarcoma PDX models, 3 hours post injection;
  • Fig. 12 A shows tumor growth over time in mice with sarcoma Sarc4809 PDX tumors treated with vehicle, cold compound nat Lu-3BP-3554, 30 MBq 177 Lu-3BP ⁇ 3554, or 60 MBq 177 Lii-3BP-
  • Fig. 12 B shows body vu s it ⁇ i ⁇ ;n over time in mice with sarcoma Sarc4809 PDX tumors treated with vehicle, cold compound nat l 3 ⁇ 4 ⁇ ' ⁇ U'-3554, 30 MBq 177 Lu-3BP-3554, or 60 MBq , 77 LU-3BP-3554.
  • 4PL means four parameter logistic curve fitting
  • ACN means acetonitrile
  • Ahx 6-Aminohexanoic acid
  • AMC means 7-amino-4-methylcoumarin amu means atomic mass unit aq. means aqueous
  • AUCinf means area under the curve extrapolated to infinity
  • BSA bovine serum albumin
  • CAF means cancer associated fibroblasts
  • CT means computed tomography
  • Cy5 means Cyanine-5
  • DAD means Diode Array Detector
  • Dde means N-(l-(4,4-dimethyl-2,6-dioxocyclohexylidene)ethyl)
  • DEG means di ethylene glycol dimethacrylate
  • DIG means N,N'-Diisopropylcarbodiimide
  • DICOM Digital Imaging and Communications in Medicine
  • DIPEA diisopropylethylamine
  • DMSO dimethyl sulfoxide
  • DOTA means 1 ,4,7, 10-tetraazacyclododecane- 1 ,4,7, 10-tetraacetic acid
  • DOTA(tBu)3-OH means Tri-terf-butyl- 1 ,4,7, 10-tetraazacyclo-dodecane- 1 ,4,7, 10-tetraacetate
  • DPP means dipeptidyl peptidase
  • ECso means half-maximal excitatory concentration
  • ECACC means European Collection of Authenticated Cell Cultures
  • EDC means 1 -ethyl-3 -(3 -dimethylaminopropyl)carbodiimide
  • EMEM Eagle's Minimum Essential Medium
  • ESI electrospray ionization
  • FACS means fluorescence-activated cell sorting
  • FAP means fibroblast activation protein
  • Fb means background fluorescent intensity
  • FBS means fetal bovine serum
  • FGF21 means fibroblast growth factor 21
  • FITC means 5(6)-fluorescein isothiocyanate
  • Fmoc 9-Fluorenylmethoxycarbonyl
  • FRET Fluorescence Resonance Energy Transfer
  • Ft fluorescent intensity
  • Gab means gamma-amino butyric acid
  • GABA means gamma-amino butyric acid
  • HATU means 0-(7-azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate
  • HBST means SPR running buffer
  • HEK-FAP means human embryonic kidney 293 cells expressing human FAP
  • HEPES means 4-(2-hydroxyethyl)- 1 -piperazineethanesulfonic acid
  • HFIP means hexafluoro-2-isopanol
  • HO Ac means acetic acid
  • HOAt means 1 -Hydroxy-7-azabenzotriazole
  • HPLC means high performance liquid chromatography
  • HPLC/MS means high performance liquid chromatography/ mass spectrometry
  • IC50 means half-maximal inhibitory concentration
  • ID/g means injected dose per gram
  • iTLC-SG means instant thin layer chromatography-silica-gel
  • K2EDTA means ethylenediaminetetraacetic acid dipotassium
  • KD dissociation constant
  • Ki inhibitory constant
  • LC/TOF-MS means Liquid chromatograpfay/time-of-flight/mass spectrometry
  • LC-MS means high performance liquid chromatography coupled with mass spectrometry
  • LDH lactate dehydrogenase
  • Leu leucine
  • LiOH means lithium hydroxide
  • M means molar or mol per Liter
  • MeV means mega electron volt
  • MMP matrix metalloproteinase
  • MRM means multiple reaction monitoring
  • Mtt Methyltrityl
  • MTV mean tumor volume
  • MW means molecular weight n.d. means not determined
  • Na2SC>4 means sodium sulfate
  • NaCl sodium chloride
  • NaHC03 sodium hydrogencarbonate
  • NCA means non-compartmental analysis
  • NHS means N -Hydroxy succinimide
  • NMP means l-methyl-2-pyrrolidone
  • Oic means L-octahydroindol-2-carbonsaure
  • Pbf means 2,2,4,6,7-pentamethyl-2,3-dihydrobenzofuran-5-sulfonyl
  • PBS phosphate buffered saline
  • PDX means patient-derived xenograft
  • PET means positron emission tomography
  • pIC50 means the negative log of the IC50 value when converted to molar
  • POP prolyl oligopeptidase
  • PREP prolyl endopeptidase
  • PS means polystyrene
  • Q-TOF means quadrupole time of flight
  • Ref reference RFU means relative fluorescence unit
  • RLB means radioligand binding assay
  • RMCE means recombinase-mediated cassette exchange
  • R t means retention time
  • RU means resonance units
  • SAR means structure activity relationship
  • SCK means single cycle kinetics
  • SPECT single photon emission computed tomography
  • SPPS Solid Phase Peptide Synthesis
  • tBu means tert. butyl
  • TFA means trifluoroacetate or trifluoroacetic acid
  • TGI tumor growth inhibition
  • THF Tetrahydrofuran
  • TIPS means triisopropyl silane
  • TLC thin layer chromatography
  • TME tumor microenvironment
  • t R retention time
  • UHPLC means ultrahigh performance liquid chromatography
  • UV means ultraviolet
  • Vss means volume of distribution at steady state
  • Vz means volume of distribution in the terminal phase
  • Solvents were used in the specified quality without further purification.
  • Acetonitrile Super Gradient, HPLC, VWR - for analytical purposes; PrepSolv, Merck - for preparative purposes); dichloromethane (synthesis, Roth); ethyl acetate (synthesis grade, Roth);
  • N,N-dimethylformamide (peptide synthesis grade, Biosolve); 1 -methyl-2-pyrolidone (peptide gradt Tech) 1 ,4-dioxane (reinst, Roth); methanol (p. a., Merck).
  • HT29 (ECACC Cat, No. 91072201) and WI-38 (ECACC Cat. No. 90020107) were purchased from ECACC and HEK293 cells expressing human FAP (Q12884) were produced by InSCREENeX GmbH (Braunschweig, Germany) using recombinase-mediated cassette exchange (RMCE).
  • RMCE recombinase-mediated cassette exchange
  • HPLC/MS analyses were performed by injection of 5 m ⁇ of a solution of the sample, using a 2 step gradient for all chromatograms (5-65% B in 12 min, followed by 65-90% in 0.5 min, A: 0.1% TFA in water and B: 0.1% TFA in ACN).
  • the‘Find Compounds by Formula’ -feature was used.
  • the individual‘neutral mass of a compound (in units of Daltons)’-values and the corresponding isotope distribution pattern were used to confirm compound identity.
  • the accuracy of the mass spectrometer was approx. ⁇ 5 ppm.
  • Preparative HPLC separations were done with reversed phase columns (Kinetex 5p XB-C18 100 A, 150 x 30 mm from Phenomenex or RLRP-S 8p, 100 A, 150 x 25 mm) as stationary phase.
  • mobile phase 0.1% TFA in water (A) and 0.1 % TFA in ACN (B) were used which were mixed in linear binary gradients.
  • the gradients are described as:“10 to 40% B in 30 min”, which means a linear gradient from 10% B (and correspondingly 90% A) to 40% B (and correspondingly 60% A) was run within 30min.
  • Flow-rates were within the range of 30 to 50 ml/min.
  • a typical gradient for the purification of the compounds of the invention started at 5-25% B and ended after 30 min at 35-50% B and the difference between the percentage B at end and start was at least 10%.
  • a commonly used gradient was“15 to 40% B in 30 min”.
  • Solid-phase synthesis was performed on polystyrene (cross linked with 1 ,4-divinylbenzene (PS) or di (ethylene glycol) dimethacrylate (DEG)), ChemMatrix (CM) or TentaGel (TG) resin.
  • Resin linkers were trityl, wang and rink amide.
  • the attachment of the first building block was performed as follows.
  • the resin polystyrene (PS) trityl chloride, initial loading: 1.8 mmol/g
  • DCM dimethyl methacrylate
  • PS polystyrene
  • the resin was washed with DCM and then treated with HF1P/DCM (7/3, 4 - 6 ml, 4 hours) and subsequently washed with DCM (3 ml, 3x 1 minute), DMF (3 ml, ml, 1 minute).
  • Coupling Coupling of building blocks/amino acids (chain assembly):
  • Terminal acetylation After additi i m I ⁇ i*‘ 3 solution (1.75 ml, 16 eq.) and acetic anhydride solution (1 75 ml, 13 eq.) the resin was shaken for 10 minutes. Afterwards the resin was washed with DMF (3 ml,
  • Cleavage method A Cleavage of protected fragments from hyper-acid labile resin:
  • the resin was finally washed with DCM (3 ml, 4x 1 minute) and then dried in the vacuum. Then the resin was treated with HFIP/DCM (7/1, 4 ml, 4 hours) and the collected solution evaporated to dryness. The residue was purified with preparative HPLC or used without further purification.
  • Cleavage method B Cleavage of unprotected fragments (complete resin cleavage):
  • the resin was finally washed with DCM (3 ml, 4x 1 minute), dried in the vacuum overnight a ft ⁇ . ited with TFA, i r ' water and TIPS (94/2.5/2.5/1) for 2 h (unless otherwise stated). Afterwards the cleavage solution was poured into a chilled mixture of MTBE and cyclohexane (1/1, 10- fold excess compared to the volume of cleavage solution), centrifuged at 4 °C for 5 minutes and the precipitate collected and dried in the vacuum. The residue was lyophilized from water/ acetonitrile prior to purification or further modification.
  • the protected/partially protected compound was dissolv water and TIPS (95/2.5/2.5) for 2 h (unless otherwise stated). Afterwards the cleavage solution was poured into a chilled mixture of MTBE and cyclohexa excess compared to the volume of cleavage solution), centrifuged at 4 °C for 5 minutes and the precipitate collected and dried in the vacuum. The residue was lyophilized from water/acetonitrile prior to purification or further modification.
  • One general synthesis route for compounds of the invention comprises
  • SPPS Solid Phase Peptide Synthesis
  • Example 2a The synthesis of the title compound was either performed by initially synthesizing the linear peptide precursor on solid phase and by subsequent solution phase cyclizations (Example 2a, either in non-aqueous solution (Method A) or in aqueous solution (Method B)) or alternatively by performing all steps on solid phase including a solid phase cyclization (Example 2b).
  • the crude peptide (based on 50 mhio ⁇ resin loading) was dissolved in 10 ml of a 1:1 mixture of ethanol and acetonitrile. To this mixture first and then 23.7 mg of 1,3,5- tris(bromomethyl)benzene (66.6 pmol, 1.3 eq compared to initial resin loading) were added. The solution was stirred for 1 hour and then 42.8 mg cysteamine (555 pmol, 11 eq compared to initial resin loading) were added. After 1 hour the solvents was removed in vacuo and 25 ml of a 1:1 mixture of acetonitrile and water (containing were added. The solvents were removed by lyophilization. The remainder was subjected to HPLC purification (15 to 45%
  • Example 2b Synthesis including solid phase cyclization method
  • the resin was washed with DC ’ I N IF, 0.9 M DIPEA in DM ⁇ ! ' E, DCM (3/3/2/3/S) and dried in the vaccum. The following cyclization was performed in 200 pmol portions. To this end, the resin was swollen in DMF and then treated with a solution of 1 ,3 , 5 -T ris(bromomethyl)benzene (86 mg, 240 pmol, 1.2 eq), DIPEA ( mmol, 5 eq) in
  • the crude peptide (based on 50 pmol resin loading) was dissolved in 10 ml of a 1 :1 mixture of ethanol and acetonitrile. To this mixture first 30 m ⁇ DIPEA and then 26.8 mg of 1 ,3,5- tris(bromomethyl)benzene (75 mhio ⁇ , 1.5 eq compared to initial resin loading) were added. After stirring the solution for 45 minutes a solution of 43 mg piperazine (500 pmol, compared loading) in 200 pi of a 1 : 1 mixture of etlianol/acetonitrile was added.
  • Condition B • at room temperature overnight (also referred to herein as Condition B) (in case ofEu(III) complexes).
  • Varian Bondesil-ENV was placed in a 15 ml polystyrene syringe, pre- washed with methanol (1 x 5 ml) and water (2 x 5 ml). Then the reaction solution was applied to the column. Thereafter elution was performed with water (2 x 5 ml - to remove excess salt), 5 ml of 50% ACN in water as first fraction and each of the next fractions were eluted with 5 ml of 50% ACN in water containing 0.1% TFA.
  • the complex was prepared starting from 25 mg peptide 3BP-3407 (15.7 pmtil) dissolved in Buffer A, diluted with a solution of Ga(NOs)3 x H2O which was treated with Condition A.
  • Purification A was employed (15 to 40% B in 30 min - RLRP-S) to yield 16.78 mg of the pure title compound (69.3% yield).
  • LC/TOF-MS exact mass 1658.664 (calculated 1658.639).
  • CTsHiosGaNieCfeoSi 1660.568).
  • the complex was prepared starting from 25 mg peptide 3BP-3407 (15.7 pmol) dissolved in Buffer A, diluted with a solution of LuCb which was treated with Condition A. In the purification step‘Purification A’ was employed (15 to 40% B in 30 min - RLRP-S) to yield 16.66 mg of the pure title compound (60.1% yield).
  • the complex was prepared starting from 9.5 mg peptide (6 pmol) 3BP-3407 dissolved in Buffer B, diluted with a solution of EuCh x 6 H 2 0 which was treated with Condition B. In the purification step‘Purification B’ was employed to yield 8.24 mg of the pure title compound (79.3% yield).
  • HPLC: R, 5.7 min.
  • the complex was prepared starting from 18 mg peptide 3BP-3554 (12.2 mitio ⁇ ) dissolved in Buffer A, diluted with a solution of Cu(OAc)2 which was treated with Condition A. In the purification step‘Purification B’ was employed to yield 16.5 mg of the pure title compound (88% yield).
  • plasma stability assay measures degradation of compounds of the present invention in blood plasma. This is an important characteristic of a compound as compounds, with the exception of pro-drugs, which rapidly degrade in plasma, generally show poor in vivo efficacy. The results show that those compounds are highly stable in human and mouse plasma. The stability is sufficient for the diagnostic, therapeutic and theragnostic use of these compounds according to the present invention.
  • the plasma stability samples were prepared by spiking 50 m ⁇ plasma aliquots (all K2EDTA) with 1 m ⁇ compound stock solution in DMSO. After vortexing the samples were incubated in a Thermomixer at 37°C for 0, 4 and 24 hours. After incubation the samples were stored on ice until further treatment. All samples were prepared in duplicates.
  • the determination of the analyte in the clean sample solutions was performed on an Agilent 1290 UHPLC system coupled to an Agilent 6530 Q-TOF mass spectrometer.
  • the chromatographic separation was carried out on a Phenomenex BioZen XB-C18 HPLC column (50 x 2 mm, 1 7 mhi particle size) with gradient elution using a mixture of 0.1% formic acid in water as eluent A and acetonitrile as elue B to 41% in 7 min, 800 m ⁇ /min, 40°C).
  • Mass spectrometric detection was performed in positive ion ESI inode by scanning the mass range from m/z 50 to 3000 with a sampling rate of 2 / sec.
  • the ion currents for the double or triple charged monoisotopic signal was extracted for both, the compound and the internal standard.
  • Quantitation was performed by external matrix calibration with internal standard using the integrated analyte signals.
  • recovery was determined by spiking a pure plasma sample that only contained the internal standard after treatment with a certain amount of the compound.
  • Carry-over was evaluated by analysis of a blank sample (20% acetonitrile) after the highest calibration sample.
  • FAP-expressing human WI-38 fibroblasts were cultured in EMEM including 15% fetal bovine serum, 2mM L-Glutamine and 1% Non-essential amino acids. Cells were detached with Accutase (Biolegend, #BLD-423201) and washed in FACS buffer (PBS including 1% FBS). Cells were diluted in FACS buffer to a final concentration of 100.000 cells per ml and 200 m ⁇ of the cell suspension are transferred to a u-shaped non-binding 96-well plate (Greiner).
  • Recombinant human FAP (R&D systems, # 3715-SE) was diluted in assay buffer (50 iiiM Tris, 1 M NaCl, 1 mg/mL BSA, pH 7.5) to a concentration of 3.6 nM. 25 pi of i lution was mixed with 25 m ⁇ of a 3-fold serial dilution of the test compounds and incubated for 5 min in a white 96-well ProxiPlate (Perkin Elmer). As specific FAP substrate the FRET-peptide HiLyteFluorTM 488 - VS(D-)P SQG K(QXL® 520) - NH2 was used (Bainbridge, et al, Sci Rep, 2017, 7: 12524).
  • the compounds of the present invention show surprisingly superior results in both the FACS Binding assay and the FAP protease activity assay.
  • Table 6 Compound ID, sequence, exact calculated mass, exact mass found, retention time in minutes as determined by HPLC and pIC50 category of FACS binding and FAP activity assay
  • a background response is generated due to the difference in the refractive indices of the running and sample buffers, as well as unspecific binding of the test compounds to the flow cell surface.
  • This background is measured and subtracted by running the sample on a control flow cell coated with the same density of capture antibody in the absence of immobilize ⁇ rthermore, baseline drift correction of the binding data is performed, which is caused by slow dissociation of the captured the immobilized antibody. This drift is measured by injecting running buffer through a flow cell with the antibody and FAP immobilized to the sensor surface.
  • BiacoreTM CMS sensor chips were used. Human anti-FAP antibody (MAE systems) was diluted in 10 mM acetate buffer, pH 4.5, to a final concentration of 50 pg/mL.
  • a 150 pL aliquot was transferred into plastic vials and placed into the sample rack of the BiacoreTM T200 instrument.
  • Amine Coupling Kit Reagent solutions were transferred into plastic vials and placed into the sample rack: 90 pL of 0.4 M l-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC), and 90 pL of 0.1 M N-hydroxysuccinimide (NHS).
  • EDC l-ethyl-3-(3-dimethylaminopropyl)carbodiimide
  • NHS N-hydroxysuccinimide
  • the BiacoreTM liquid system was set-up as follows: Separate bottles containing distilled water (1 L), Running Buffer (500 mL), as well as an empty bottle for waste were placed onto the buffer tray. A preinstalled program for immobilization was used, with an immobilization level of 7000 RU. Immobilization was performed at 25°C. The immobilization procedure of anti-FAP antibodies was performed, as described in the Table 7.
  • Human recombinar was diluted in Running Buffer to a final concentration of 20 pg/mL. A 100 pL aliquot of human Vorking-Solution was transferred into plastic vials and placed into a sample rack. A 0.5 mM Compound-Stock-Solution was prepared by dissolving each compound in DMSO. For each test compound, Compound-Stock-Solutions were diluted in Running Buffer (HBST) at 500 nM and further diluted with HBST-DMSO Buffer (0.1 % DMSO). SPR binding analyses for binary complexes were performed in SCK mode at 25°C. Table 8 describes the protocol for capturing and assessment of the binding kinetics. Following three SCK measurements, a baseline drift was assessed by injecting running buffer through a flow cell, with the antibody and FAP immobilized to the sensor surface.
  • HBST Running Buffer
  • HBST-DMSO Buffer 0.1 % DMSO
  • protease activity assays were performed analogues to otivity assay described above with following exceptions.
  • PREP activity was measured with recombinant human PREP (R&D systems, #4308-SE).
  • R&D systems #4308-SE
  • As substrate 50 pM Z-GP-AMC (Bachem, # 4002518) was used.
  • the DPP4 activity assay was performed in DPP assay buffer (25 mM Tris, pH 8.0).
  • Recombinant human DPP4 was purchased from R&D systems (#9168-SE). 20 mM of GP-AMC (Santa Cruz Biotechnology, #115035-46-6) was used as substrate.
  • the specificity screening was carried out in order to early identify significant off-target interactions of compounds of the present invention.
  • the specificity was tested using a standard battery of assays (“SafetyScreen44TM Panel 44 ) comprising 44 selected targets and compounds binding thereto (referred to as“reference compounds”, Ref. Compounds), recommended by Bowes et al. (Bowes, et at, Nat Rev Drug Discov, 2012, 11: 909).
  • the reference compounds served as positive controls for the respective assays, therefore inhibition is expected to be detected with these reference compounds.
  • the compounds of the invention were not expected to show inhibition in these assays.
  • These binding and enzyme inhibition assays were performed by Eurofins Cerep SA (Celle TEvescault, France).
  • 3BP-3407 and 3BP-3554 were tested at IOmM Compound binding was calculated as % inhibition of the binding of a radioactively labeled ligand specific for each target (“% Inhibition of Specific Binding” (3BP-3407) or (3BP-3554), respectively. Compound enzyme inhibition effect was calculated as % inhibition of control enzyme activity.
  • Results showing an inhibition or stimulation higher than 50% are considered to represent significant effects of the test compounds. Such effects were not observed at any of the receptors studied which are listed in the following Table 1 1.
  • Table 11 Results of the specificity screening (SafetyScreen44TM Panel) for 10 mM 3BP-3407 and 10 mM 3BP-3554
  • 3BP-3407 and 3BP-3554 were tested at 1 mM and 10 mM in duplicates.
  • the fluorescent intensity (Ft) in each data set was defined as 100% activity.
  • the background fluorescent intensil ach data set was defined as 0% activity.
  • Table 12 Results of the specificity protease screening for 1 mM and 10 mM 3BP-3407 and 1 mM and 10 mM 3BP-3554
  • Example 11 m In- and ,77 Lu-labeling of selected compounds
  • a compound In order to serve as a diagnostically, therapeutically, or theragnostically active agent, a compound needs to be labeled with a radioactive isotope.
  • the labeling procedure needs to be appropriate to ensure a high radiochemical yield and purity of the radiolabeled compound of the invention.
  • This example shows that the compounds of the present invention are appropriate for radiolabeling and can be labeled in high radiochemical yield and purity.
  • 30-100 MBq of 111 M3 ⁇ 4 (in 0.02 M HC1) were mixed with 1 nmol of compound (200 mM stock solution in 0.1 M HEPES pH 7) per 30 MBq and buffer (1 M sodium acetate buffer pH 5 or 1 M sodium acetate / ascorbic acid buffer pH 5 containing 25 mg/ l methionine) at a final buffer concentration of 0.1 - 0.2 M.
  • the mixture was heated to 80 °C for 20-30 min. After cooling down, and TWEEN-20 were added at a final concentration of 0.2 mM and 0.1 %, respectively.
  • 0.2 - 2.0 GBq 177 LuCl3 (in 0.04 M HC1) were mixed with 1 nmol of compound (200 mM stock solution in 0.1 M HEPES pH 7) per 45 MBq and buffer (1 M sodium acetate / ascorbic acid buffer pH 5 containing 25 mg/ml methionine) at a final buffer concentration of -0.4 3 ⁇ 4 mixture was heated to 90 °C for 20 min.
  • the labeling efficiency was analyzed by thin layer chromatography (TLC) and HPLC.
  • TLC thin layer chromatography
  • HPLC HPLC
  • 1-2 pi of diluted labeling solution was applied to a strip of iTLC-SG chromatography paper (Agilent, 7.6 x 2.3 mm) and developed in citrate-dextrose solution (Sigma).
  • the iTLC strip was then cut into 3 pieces and associated radioactivity was measured with a gamma- counter.
  • the radioactivity measured at the solvent front represents free radionuclide and colloids, whereas the radioactivity at the origin represents radiolabeled compound.
  • HPLC 5 pi of diluted labeling solution was analyzed with a Poroshell SB-C18 2,7 pin (Agilent).
  • Eluent A MeCN, eluent B: H2O, 0.1 % TFA, gradient from 5% B to 70% B within 15 min, flow rate 0.5 ml/min; detector: Nal (Raytest), DAD 230 nm.
  • the peak eluting with the dead volume represents free radionuclide, the peak eluting with the peptide-specific retention time as determined with an unlabeled sample represents radiolabeled compound.
  • Radionuclidic incorporation yield was > 95% and radiochemical purity > 90% at end of synthesis.
  • Exemplary radiochemical purities for m In-labeled compounds are shown in Table 13.
  • 177 Lu-labeled compounds in formulations suitable for human use maintained a radiochemical purity of > 90% up to 6 days post synthesis (Table 14).
  • the radiochromatograms for selected compounds are shown in Figs. 1 to 4, whereby Fig. 1 shows a radiochromatogramm of 177 LU-3BP-3407 in formulation buffer containing 100 mg/mL ascorbate and 5 mg/mL L- methionine analyzed immediately upon end of synthesis, Fig.
  • Fig. 2 shows a radiochromatogramm of 177 LU-3BP-3407 in formulation buffer containing 100 mg/mL ascorbate and 5 mg/mL L- methionine analyzed six days post end of synthesis
  • Fig. 3 shows a radiochromatogramm of 177 LU-3BP-3554 in formulation buffer containing 100 mg/mL ascorbate and 5 mg/mL L- methionine analyzed immediately upon end of synthesis
  • Fig, 4 shows a radiochromatogram of 177 Lu-3BP-3554 in formulation buffer containing 100 mg/mL ascorbate and 5 mg/mL L-methionine analyzed six days post end of synthesis.
  • Table 14 Radiochemical purity by HPLC of 177 Lu-labeled compounds in a formulation buffer containing 100 mg/mL ascorbate and 5 mg/mL L-methionine analyzed on day 0 and day 6 post end of synthesis.
  • Example 12 Imaging ami biodistribution studies
  • Radioactively labeled compounds can be detected by imaging methods such as SPECT and PET, Furthermore, the data acquired by such techniques can be confirmed by direct measurement of radioactivity contained in the individual organs prepared from an animal injected with a radioactively labeled compound of the invention. Thus, the biodistribution (the measurement of radioactivity in individual organs) of a radioactively labeled compound can be determined and analyzed. This example shows that the compounds of the present invention show a biodistribution appropriate for diagnostic imaging and therapeutic treatment of tumors.
  • mice were inoculated with 5 x 10 6 HEK-FAP (embryonic human kidney 293 cells genetically engineered to express high levels of FAP) cells in one shoulder.
  • HEK-FAP embryonic human kidney 293 cells genetically engineered to express high levels of FAP
  • mice received ⁇ 30 MBq 1 1 fin-labelled compounds of the invention (diluted to 100 pL with PBS) administered intravenously via the tail vein.
  • Images were obtained on a NanoSPECT/CT system (Mediso Medical Imaging Systems, Budapest, Hungary) using exemplarily the following acquisition and reconstruction parameters (Table 15).
  • Imaging data were saved as DICOM files and analysed using VivoQuantTM software (Invicro, Boston, USA). Results are expressed as a percentage of injected dose per gram of tissue (%ID/g).
  • %ID/g For biodistribution studies, animals were sacrificed by cervical dislocation at 24h or 48h post injection and then dissected. Different organs and tissues were collected and weighed, and the radioactivity was determined by g-counting. Two animals were used per time point. Results are expressed as a percentage of injected dose per gram of tissue (%ID/g).
  • Radioactively labeled compounds can be used for therapeutic and diagnostic application in various diseases, especially cancer.
  • This example shows that the compounds of the present invention have anti-tumor activity suitable for the therapeutic treatment of tumors.
  • mice Female swiss nude mice (7- to 8-week-old, Charles River Laboratories, France) were inoculated with 5 x 10 6 HEK-FAP cells in one shoulder, and treatments were administered when the tumors reached a mean tumor volume of of 160 ⁇ 44 mm 3 . Mice were divided into 4 different groups of 10 animals/group: Group 1 - vehicle control, Group 2 - cold compound nat Lu-3BP- 3554, Group 3 - 30 MBq 177 Lu-3BP-3554 (low dose), and Group 4 - 60 MBq Lu-FAP-3554 (high dose).
  • Treatments were administered on Day 0 by intravenous injection into the tail vein at 4 mL/kg (100 pL/mouse). Tumor volume and body weights were measured on Day 0 (i.e. the first day of radiotracer administration) and then thrice weekly until completion of the study.
  • mice injected with Lu-labeled 3BP-3554 were determined by SPECT imaging in three mice per dosing group. Subsequently, following SPECT, a CT scan was done for anatomical information. Imaging was performed 3 h, 24 h, 48 h and 120 h post injection with a NanoSPECT/CT system (Mediso Medical Imaging Systems, Budapest, Hungary) using exemplarily the following acquisition and reconstruction parameters (Table 16).
  • Imaging data were saved as DICOM files and analysed using VivoQuantTM software (Invicro, Boston, USA). Results are expressed as a percentage of injected dose per gram of tissue (%ID/g),
  • Tumors in vehicle and cold compound nat Lu-3BP-3554- treated mice reached a mean tumor volume (MTV) of 1338 ⁇ 670 mm 3 and 1392 ⁇ 420 mm 3 on day 14, respectively (Fig. 9 A).
  • MTV mean tumor volume
  • Statistically significant (P ⁇ 0.01) anti-tumor activity was observed in mice of both treatment groups.
  • Tumor growth inhibition (TGI) at day 14 was 11 1% and 113% in mice treated with a single dose of 30 or 60 MBq 177 Lu-3BP-3554, respectively, relative to the vehicle-treated group.
  • the MTV in all mice treated with 177 Lu-3 BP-3554 was reduced to ⁇ 70 mm 3 on day 14.
  • SPECT/CT imaging of 3 animals of both l 77 Lu-labeled treatment groups showed high tumor- to-background contrast during all examined time points (3-120 h post-injection (pi.)). High tumor retention up to 120 h was observed.
  • the organ with the highest non-target uptake was the kidney, with tumor-to-kidney ratios of 8.6 ⁇ 0.6 and 8.0 ⁇ 1.6 at 3 h pi. m iui « treated with 30 or 60 MBq l 77 Lu-3BP-3554, respectively. These ratios increased over time, attaining the highest value at 120 h with 40 ⁇ 7.9 and 32 ⁇ 7.4 tumor-to-kidney ratios in mice treated with 30 or 60 MBq 177 Lu-3BP ⁇ 3554, respectively.
  • An exemplary panel of SPECT/CT images for mouse 5 which is a high-dose animal is shown in Fig. 10 A and for mouse 1 which is a low-dose animal is shown i
  • the Sarc4183, Sarc4605, Sarc4809 and Sard 2616 PDX models were derived from patients with rhabdomyosarcoma, osteosarcoma, undifferentiated sarcoma and undifferentiated pleiomorphic sarcoma, respectivt perimental Pharmacology & Oncology Berlin-Buch, Germany). Tumor fragments were transplanted subcutaneously in the left flank of 8-week-old NMRI nu/nu mice (Janvier Labs, France). All animal experiments were conducted in compliance with the German animal protection laws. 47 days (5arc41S3, Sarc4809) or 46 days (Sarc4605, Sar 2616) after transplantation, 2-3 mice per model were imaged 3 hours after a single intravenous injection of 30 MBq of 111 ] Imaging was performed as described in Example 12.
  • Fig. 11 A Quantification of tumor uptake of two (Sarc4605, Sard 2616) or three (Sarc4183, Sarc4809) PDX-bearing mice, respectively, revealed %ID/g values of 4.9 ⁇ 1.7 (Sarc4183), 5.2 ⁇ 0.8 (Sarc4605), 4.4 ⁇ 0.7 (Sarc4809) and 6.1 ⁇ 0.6 (Sard 2616) as shown in Fig. 11 B.
  • These results demonstrate i n In-3BP-3554 uptake in all 4 sarcoma models. Tumor-to-kidney ratios were 4.7 ⁇ 1.2 (Sarc4183), 3.2 ⁇ 0.4 (Sarc4605) 4.1 ⁇ 0.7 (Sarc4809) and 4.3 ⁇ 1.2 (Sarcl2616).
  • mice All tumors continuously grew throughout the follow-up period of the study until day 42.
  • Tumors in vehicle and nat Lu-3BP-3554 treated mice reached an MTV of 894 ⁇ 610 mm 3 and 1225 ⁇ 775 mm 3 on day 31 (the last day on which at least 50% mice per group were still alive), respectively.
  • Tumors in mice treated with a single dose of 30 or 60 MBq 177 Lu- 3BP-3554 reached an MTV of 635 ⁇ 462 and 723 ⁇ 391 mm 3 on day 31, respectively (Fig. 12A).
  • Statistically significant (P ⁇ 0.05) anti-tumor activity was observed in mice of both treatment groups.
  • TGI Tumor growth inhibition
  • mice and rats The pharmacokinetic behavior of selected compounds was assessed in mice and rats. This characterization of the pharmacokinetic behavior of a compound enables new insights into distribution arid elimination of the compound and the calculation of the exposure.
  • the formulations were applied intravenous with a dose of 4 nmol/kg, 40 mnol/kg and 400 nmol/kg in mice and 2 nmo/kg, 20 nmol/kg and 200 nmol/kg (3BP-3554) or 40 nmol/kg and 400 mol/kg (3 BP-3623) in rats. Assuming an allometric translation factor of 12.3 from human to mouse, and 6.2 from human to rats (Nair AB, Jacob S. Journal of Basic and Clinical Pharmacy, 2016, 7(2): 27-31), the applied doses represent a human dose range of 0.325 nmol/kg to 32.5 nmol/kg.
  • the compounds were quantified in the prepared plasma samples were subjected to a protein precipitation procedure.
  • the determination of the analyte in the clean sample solutions was performed on an Agilent 1290 UHPLC system coupled to an Agilent 6470 triple quadrupole mass spectrometer.
  • the chromatographic separation was carried out on a Pfaeiionienex BioZen Peptide XB-C18 HPLC column (50 x 2 mm, 1.7 p particle size) at 40°C with gradient elution using a mixture of0.1% formic acid in water as eluent A and acetonitrile as eluent B (isocratic at 5% B for 1 min followed by a linear gradient to 43% B in 4 min, 500 m ⁇ /min).
  • Mass spectrometric detection was performed in positive ion ESI mode by multiple reaction monitoring (MRM). Table 17: Mass spectrometric detection parameters
  • Quantitation of test items was accomplished using the Quantitative Analysis software of the Agilent MassHunter software suite. A quadratic regression was performed with a weighting factor of 1/x.
  • NCA non-compartmental analysis
  • Table 21 Summary of NCA parameters of 3BP-3623 in rat plasma

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Optics & Photonics (AREA)
  • Physics & Mathematics (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Peptides Or Proteins (AREA)
EP20735646.0A 2019-07-08 2020-07-08 Compounds comprising a fibroblast activation protein ligand and use thereof Pending EP3997103A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP19000325.1A EP3763726A1 (en) 2019-07-08 2019-07-08 Compounds comprising a fibroblast activation protein ligand and use thereof
EP19198813 2019-09-20
PCT/EP2020/069298 WO2021005125A1 (en) 2019-07-08 2020-07-08 Compounds comprising a fibroblast activation protein ligand and use thereof

Publications (1)

Publication Number Publication Date
EP3997103A1 true EP3997103A1 (en) 2022-05-18

Family

ID=71409439

Family Applications (1)

Application Number Title Priority Date Filing Date
EP20735646.0A Pending EP3997103A1 (en) 2019-07-08 2020-07-08 Compounds comprising a fibroblast activation protein ligand and use thereof

Country Status (12)

Country Link
US (1) US20220273831A1 (zh)
EP (1) EP3997103A1 (zh)
JP (1) JP2022541753A (zh)
KR (1) KR20220032079A (zh)
CN (1) CN114341159B (zh)
AU (1) AU2020310538A1 (zh)
BR (1) BR112022000122A2 (zh)
CA (1) CA3145340A1 (zh)
CL (1) CL2021003580A1 (zh)
IL (1) IL289675A (zh)
MX (1) MX2022000252A (zh)
WO (1) WO2021005125A1 (zh)

Families Citing this family (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3176497A1 (en) 2020-05-07 2021-11-11 Fatima MECHTA-GRIGORIOU Antxr1 as a biomarker of immunosuppressive fibroblast populations and its use for predicting response to immunotherapy
WO2022160338A1 (zh) * 2021-02-01 2022-08-04 五一一制药股份有限公司 成纤维细胞活化蛋白-α显像剂的放射性标记配体及其制备方法
CN114369084B (zh) * 2021-02-10 2023-02-03 烟台蓝纳成生物技术有限公司 一种截短型伊文思蓝修饰的成纤维细胞活化蛋白抑制剂及其制备方法和应用
AU2021427618B2 (en) 2021-02-10 2023-08-17 Yantai Lannacheng Biotechnology Co., Ltd. Truncated evans blue modified fibroblast activation protein inhibitor, preparation method therefor, and application thereof
GB202109922D0 (en) 2021-07-09 2021-08-25 Blue Earth Diagnostics Ltd Radiotracers and therapeutics binding to fibroblast activation protein (fap)
WO2023152671A1 (en) 2022-02-09 2023-08-17 Novartis Ag Pharmaceutical compositions comprising a 225-actinium-labelled complex and a bismuth sequestering agent
EP4282439A1 (en) 2022-05-23 2023-11-29 Erasmus University Rotterdam Medical Center Radioisotope labeled sstr2-agonists with linkers
CN115304582B (zh) * 2022-07-20 2023-05-12 北京法伯新天医药科技有限公司 FAP-α特异性肿瘤诊断显像剂
EP4311557A1 (en) * 2022-07-26 2024-01-31 Oncomatryx Biopharma, S.L. Fap-targeted antibody-drug conjugates
WO2024052431A1 (en) * 2022-09-07 2024-03-14 3B Pharmaceuticals Gmbh Prostate specific membrane antigen (psma) ligands and use thereof
EP4342890A1 (en) 2022-09-21 2024-03-27 Erasmus University Rotterdam Medical Center Platform and scaffold for fap targeting agents

Family Cites Families (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2285785T3 (es) 1997-09-29 2007-11-16 Point Therapeutics, Inc. Estimulacion de celulas hematopoyeticas in vitro.
EP0953639A1 (en) 1998-04-30 1999-11-03 Boehringer Ingelheim International GmbH FAPalpha-specific antibody with improved producibility
US6890904B1 (en) 1999-05-25 2005-05-10 Point Therapeutics, Inc. Anti-tumor agents
AU2001256325A1 (en) 2000-03-17 2001-09-24 Boehringer Ingelheim Pharma Kg Human and humanized fap-alpha-specific antibodies
EP1452868A2 (en) * 2003-02-27 2004-09-01 Pepscan Systems B.V. Method for selecting a candidate drug compound
US7374898B2 (en) * 2004-10-12 2008-05-20 The Research Foundation Of State University Of New York Peptide inhibitors against seprase
JP2008545661A (ja) 2005-05-19 2008-12-18 ジェネンテック・インコーポレーテッド 線維芽細胞活性化タンパク質阻害剤化合物および
US8933201B2 (en) 2006-06-07 2015-01-13 The Board Of Regents Of The University Of Oklahoma Substrates and inhibitors of antiplasmin cleaving enzyme and fibroblast activation protein and methods of use
AU2008228863A1 (en) 2007-03-20 2008-09-25 Trustees Of Tufts College Inhibitors of fibroblast activation protein, and methods of use thereof
US20100098633A1 (en) 2008-09-25 2010-04-22 Molecular Insight Pharmaceuticals, Inc. Selective seprase inhibitors
US8999342B2 (en) 2009-10-02 2015-04-07 Ludwig Institute For Cancer Research, Ltd. Anti-fibroblast activation protein antibodies and methods and uses thereof
NO2603530T3 (zh) 2010-08-13 2018-04-07
WO2013107820A1 (en) 2012-01-17 2013-07-25 Universiteit Antwerpen Novel fap inhibitors
EP2740726A1 (en) * 2012-12-07 2014-06-11 3B Pharmaceuticals GmbH Neurotensin receptor ligands
UA118028C2 (uk) 2013-04-03 2018-11-12 Рош Глікарт Аг Біспецифічне антитіло, специфічне щодо fap і dr5, антитіло, специфічне щодо dr5, і спосіб їх застосування
GB201402006D0 (en) 2014-02-06 2014-03-26 Oncomatryx Biopharma S L Antibody-drug conjugates and immunotoxins
EP2954933A1 (en) * 2014-06-10 2015-12-16 3B Pharmaceuticals GmbH Conjugate comprising a neurotensin receptor ligand
RU2743781C2 (ru) * 2014-06-10 2021-02-25 3Б Фармасьютикалз Гмбх Конъюгат, содержащий лиганд рецепторов нейротензина, и его применение
WO2016146174A1 (en) 2015-03-17 2016-09-22 Biontech Ag Compositions and methods for diagnosis and treatment of cancer
AU2017277463A1 (en) 2016-06-10 2019-01-03 Bayer As Radio-pharmaceutical complexes
JP2018035137A (ja) 2016-07-13 2018-03-08 マブイミューン ダイアグノスティックス エイジーMabimmune Diagnostics Ag 新規な抗線維芽細胞活性化タンパク質(fap)結合薬剤およびその使用
US20200330624A1 (en) 2017-10-23 2020-10-22 The Johns Hopkins University Imaging and radiotherapeutics agents targeting fibroblast-activation protein-alpha (fap-alpha)

Also Published As

Publication number Publication date
CA3145340A1 (en) 2021-01-14
WO2021005125A1 (en) 2021-01-14
BR112022000122A2 (pt) 2022-04-26
CL2021003580A1 (es) 2022-08-05
US20220273831A1 (en) 2022-09-01
JP2022541753A (ja) 2022-09-27
AU2020310538A1 (en) 2022-01-27
MX2022000252A (es) 2022-02-21
CN114341159A (zh) 2022-04-12
IL289675A (en) 2022-03-01
CN114341159B (zh) 2024-04-26
KR20220032079A (ko) 2022-03-15

Similar Documents

Publication Publication Date Title
EP3997103A1 (en) Compounds comprising a fibroblast activation protein ligand and use thereof
US10745445B2 (en) Conjugates that are configured for targeted delivery of therapeutic compounds to senescent cells
US20220315554A1 (en) Compounds comprising a fibroblast activation protein ligand and use thereof
US20240115745A1 (en) Compounds comprising a fibroblast activation protein ligand and use thereof
EP3763726A1 (en) Compounds comprising a fibroblast activation protein ligand and use thereof
WO2023002045A1 (en) Fibroblast activation protein inhibitors and use thereof
WO2022123462A1 (en) Radiolabelled prostate specific membrane antigen (psma) inhibitors and use thereof
US20230212549A1 (en) Compounds comprising a fibroblast activation protein ligand and use thereof
WO2023111350A2 (en) Carbonic anhydrase ix ligands
TW202334177A (zh) 包含纖維母細胞活化蛋白配體之化合物和彼之用途
WO2024052431A1 (en) Prostate specific membrane antigen (psma) ligands and use thereof

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: UNKNOWN

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20220202

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40065560

Country of ref document: HK

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20230120

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

INTC Intention to grant announced (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20231005

GRAJ Information related to disapproval of communication of intention to grant by the applicant or resumption of examination proceedings by the epo deleted

Free format text: ORIGINAL CODE: EPIDOSDIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTC Intention to grant announced (deleted)
INTG Intention to grant announced

Effective date: 20240208